1
|
Sanford SL, Badstübner M, Gerber M, Mannherz W, Lampl N, Dannenberg R, Hinchie A, Schaich MA, Myong S, Hedglin M, Agarwal S, Alder JK, Stone MD, Opresko PL. Chemotherapeutic 6-thio-2'-deoxyguanosine selectively targets and inhibits telomerase by inducing a non-productive telomere-bound telomerase complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636339. [PMID: 39975053 PMCID: PMC11838547 DOI: 10.1101/2025.02.05.636339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Most cancers upregulate the telomere lengthening enzyme telomerase to achieve unlimited cell division. How chemotherapeutic nucleoside 6-thio-2'-deoxyguanosine (6-thio-dG) targets telomerase to inhibit telomere maintenance in cancer cells and tumors was unclear. Here, we demonstrate that telomerase insertion of 6-thio-dGTP prevents synthesis of additional telomeric repeats but does not disrupt telomerase binding to telomeres. Specifically, 6-thio-dG inhibits telomere extension after telomerase translocates along its product DNA to reposition the template, inducing a non-productive complex rather than enzyme dissociation. Furthermore, we provide direct evidence that 6-thio-dG treatment inhibits telomere synthesis by telomerase in cancer cells. In agreement, telomerase-expressing cancer cells harboring critically short telomeres are more sensitive to 6-thio-dG and show a greater induction of telomere losses compared to cancer cells with long telomere reserves. Our studies reveal that telomere length and telomerase status determine 6-thio-dG sensitivity and uncover the molecular mechanism by which 6-thio-dG selectively inhibits telomerase synthesis of telomeric DNA.
Collapse
Affiliation(s)
| | - Mareike Badstübner
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - William Mannherz
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Noah Lampl
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Dannenberg
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Angela Hinchie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew A Schaich
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sua Myong
- Program in Cell, Molecular, Developmental Biology and Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Hedglin
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Suneet Agarwal
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jonathan K Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D. Stone
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Patricia L Opresko
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Narang S, Ghebrechristos Y, Evensen NA, Murrell N, Jasinski S, Ostrow TH, Teachey DT, Raetz EA, Lionnet T, Witkowski M, Aifantis I, Tsirigos A, Carroll WL. Clonal evolution of the 3D chromatin landscape in patients with relapsed pediatric B-cell acute lymphoblastic leukemia. Nat Commun 2024; 15:7425. [PMID: 39198446 PMCID: PMC11358475 DOI: 10.1038/s41467-024-51492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
Relapsed pediatric B-cell acute lymphoblastic leukemia (B-ALL) remains one of the leading causes of cancer mortality in children. We performed Hi-C, ATAC-seq, and RNA-seq on 12 matched diagnosis/relapse pediatric leukemia specimens to uncover dynamic structural variants (SVs) and 3D chromatin rewiring that may contribute to relapse. While translocations are assumed to occur early in leukemogenesis and be maintained throughout progression, we discovered novel, dynamic translocations and confirmed several fusion transcripts, suggesting functional and therapeutic relevance. Genome-wide chromatin remodeling was observed at all organizational levels: A/B compartments, TAD interactivity, and chromatin loops, including some loci shared by 25% of patients. Shared changes were found to drive the expression of genes/pathways previously implicated in resistance as well as novel therapeutic candidates, two of which (ATXN1 and MN1) we functionally validated. Overall, these results demonstrate chromatin reorganization under the selective pressure of therapy and offer the potential for discovery of novel therapeutic interventions.
Collapse
Affiliation(s)
- Sonali Narang
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Yohana Ghebrechristos
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Nikki A Evensen
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Nina Murrell
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Sylwia Jasinski
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| | - Talia H Ostrow
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - David T Teachey
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth A Raetz
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| | - Timothee Lionnet
- Institute for Systems Genetics and Department of Cell Biology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Matthew Witkowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iannis Aifantis
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Department of Pathology, NYU Langone Health, New York, NY, USA.
| | - Aristotelis Tsirigos
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Department of Pathology, NYU Langone Health, New York, NY, USA.
| | - William L Carroll
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
3
|
Nguyen T, Aida T, Iijima‐Yamashita Y, Tamai M, Nagamachi A, Kagami K, Komatsu C, Kasai S, Akahane K, Goi K, Inaba T, Sanada M, Inukai T. Application of prime editing system to introduce TP53 R248Q hotspot mutation in acute lymphoblastic leukemia cell line. Cancer Sci 2024; 115:1924-1935. [PMID: 38549229 PMCID: PMC11145152 DOI: 10.1111/cas.16162] [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: 08/15/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 06/04/2024] Open
Abstract
In childhood acute lymphoblastic leukemia (ALL), TP53 gene mutation is associated with chemoresistance in a certain population of relapsed cases. To directly verify the association of TP53 gene mutation with chemoresistance of relapsed childhood ALL cases and improve their prognosis, the development of appropriate human leukemia models having TP53 mutation in the intrinsic gene is required. Here, we sought to introduce R248Q hotspot mutation into the intrinsic TP53 gene in an ALL cell line, 697, by applying a prime editing (PE) system, which is a versatile genome editing technology. The PE2 system uses an artificial fusion of nickase Cas9 and reverse-transcriptase to directly place new genetic information into a target site through a reverse transcriptase template in the prime editing guide RNA (pegRNA). Moreover, in the advanced PE3b system, single guide RNA (sgRNA) matching the edited sequence is also introduced to improve editing efficiency. The initially obtained MDM2 inhibitor-resistant PE3b-transfected subline revealed disrupted p53 transactivation activity, reduced p53 target gene expression, and acquired resistance to chemotherapeutic agents and irradiation. Although the majority of the subline acquired the designed R248Q and adjacent silent mutations, the insertion of the palindromic sequence in the scaffold hairpin structure of pegRNA and the overlap of the original genomic DNA sequence were frequently observed. Targeted next-generation sequencing reconfirmed frequent edit errors in both PE2 and PE3b-transfected 697 cells, and it revealed frequent successful edits in HEK293T cells. These observations suggest a requirement for further modification of the PE2 and PE3b systems for accurate editing in leukemic cells.
Collapse
Affiliation(s)
- Thao Nguyen
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Tomomi Aida
- McGovern Institute for Brain ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yuka Iijima‐Yamashita
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Minori Tamai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Keiko Kagami
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Chiaki Komatsu
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Shin Kasai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Koshi Akahane
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Kumiko Goi
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Toshiya Inaba
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Masashi Sanada
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Takeshi Inukai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| |
Collapse
|
4
|
Kroeze E, Weijers DD, Kleisman MM, Ilan U, Bladergroen RS, Hagelaar R, Meijerink JPP, Jongmans MCJ, Loeffen JLC, Kuiper RP. T-cell lymphoblastic lymphoma in constitutional mismatch repair deficiency (CMMRD): Exploring treatment opportunities. Hemasphere 2024; 8:e73. [PMID: 38741596 PMCID: PMC11089271 DOI: 10.1002/hem3.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Emma Kroeze
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Dilys D. Weijers
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Uri Ilan
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Rico Hagelaar
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Jules P. P. Meijerink
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Present address:
Acerta‐Pharma (AstraZeneca)OssThe Netherlands
| | - Marjolijn C. J. Jongmans
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of Genetics, Utrecht University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| | | | - Roland P. Kuiper
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of Genetics, Utrecht University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
5
|
Contreras Yametti GP, Robbins G, Chowdhury A, Narang S, Ostrow TH, Kilberg H, Greenberg J, Kramer L, Raetz E, Tsirigos A, Evensen NA, Carroll WL. SETD2 mutations do not contribute to clonal fitness in response to chemotherapy in childhood B cell acute lymphoblastic leukemia. Leuk Lymphoma 2024; 65:78-90. [PMID: 37874744 PMCID: PMC11874253 DOI: 10.1080/10428194.2023.2273752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
Mutations in genes encoding epigenetic regulators are commonly observed at relapse in B cell acute lymphoblastic leukemia (B-ALL). Loss-of-function mutations in SETD2, an H3K36 methyltransferase, have been observed in B-ALL and other cancers. Previous studies on mutated SETD2 in solid tumors and acute myelogenous leukemia support a role in promoting resistance to DNA damaging agents. We did not observe chemoresistance, an impaired DNA damage response, nor increased mutation frequency in response to thiopurines using CRISPR-mediated knockout in wild-type B-ALL cell lines. Likewise, restoration of SETD2 in cell lines with hemizygous mutations did not increase sensitivity. SETD2 mutations affected the chromatin landscape and transcriptional output that was unique to each cell line. Collectively our data does not support a role for SETD2 mutations in driving clonal evolution and relapse in B-ALL, which is consistent with the lack of enrichment of SETD2 mutations at relapse in most studies.
Collapse
Affiliation(s)
- Gloria P. Contreras Yametti
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Gabriel Robbins
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Ashfiyah Chowdhury
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Sonali Narang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Talia H. Ostrow
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Harrison Kilberg
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Joshua Greenberg
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Lindsay Kramer
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Elizabeth Raetz
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Aristotelis Tsirigos
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Nikki A. Evensen
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - William L. Carroll
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
- Department of Pathology, NYU Langone Health, New York, NY
| |
Collapse
|
6
|
Ma Y, Mu X, Gao R, Zhang Y, Geng Y, Chen X, Yin X, Li F, He J. Maternal exposure to dibutyl phthalate regulates MSH6 crotonylation to impair homologous recombination in fetal oocytes. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131540. [PMID: 37167869 DOI: 10.1016/j.jhazmat.2023.131540] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Homologous recombination (HR) during early oogenesis repairs programmed double-strand breaks (DSBs) to ensure female fertility and offspring health. The exposure of fetal ovaries to endocrine disrupting chemicals (EDCs) can cause reproductive disorders in the adulthood. The EDC dibutyl phthalate (DBP) is widely distributed in flexible plastic products, leading to ubiquitous human exposure. Here, we report that maternal exposure to DBP caused gross aberrations in meiotic prophase I of fetal oocytes, including delayed progression, impaired DNA damage response, uncoupled localization of DMC1 and RAD51, and decreased HR. However, programmed DSBs were efficiently repaired. DBP exposure negatively regulated lysine crotonylation (Kcr) of MSH6. Similar meiotic defects were observed in fetal ovaries with targeted disruption of Msh6, and mutation of K544cr of MSH6 impaired its association with Ku70, thereby promoting non-homologous end joining (NHEJ) and inhibiting HR. Unlike mature F1 females, F2 female mice exhibited premature follicular activation, precocious puberty, and anxiety-like behaviors. Therefore, DBP can influence early meiotic events, and Kcr of MSH6 may regulate preferential induction of HR or NHEJ for DNA repair during meiosis.
Collapse
Affiliation(s)
- Yidan Ma
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xinyi Mu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Rufei Gao
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan Zhang
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuemei Chen
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Yin
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Fangfang Li
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
7
|
Teng JY, Yang DP, Tang C, Fang HS, Sun HY, Xiang YN, Li XM, Yang F, Xia RX, Fan F, Liu J, Yu J, Hu JC, Li BS, Li H, Meng FL, Duan CW, Zhou BBS. Targeting DNA polymerase β elicits synthetic lethality with mismatch repair deficiency in acute lymphoblastic leukemia. Leukemia 2023:10.1038/s41375-023-01902-3. [PMID: 37095208 DOI: 10.1038/s41375-023-01902-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 03/26/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
Mismatch repair (MMR) deficiency has been linked to thiopurine resistance and hypermutation in relapsed acute lymphoblastic leukemia (ALL). However, the repair mechanism of thiopurine-induced DNA damage in the absence of MMR remains unclear. Here, we provide evidence that DNA polymerase β (POLB) of base excision repair (BER) pathway plays a critical role in the survival and thiopurine resistance of MMR-deficient ALL cells. In these aggressive resistant ALL cells, POLB depletion and its inhibitor oleanolic acid (OA) treatment result in synthetic lethality with MMR deficiency through increased cellular apurinic/apyrimidinic (AP) sites, DNA strand breaks and apoptosis. POLB depletion increases thiopurine sensitivities of resistant cells, and OA synergizes with thiopurine to kill these cells in ALL cell lines, patient-derived xenograft (PDX) cells and xenograft mouse models. Our findings suggest BER and POLB's roles in the process of repairing thiopurine-induced DNA damage in MMR-deficient ALL cells, and implicate their potentials as therapeutic targets against aggressive ALL progression.
Collapse
Affiliation(s)
- Ji-Yuan Teng
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ding-Peng Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chao Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hou-Shun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Ying Sun
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue-Ning Xiang
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Meng Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui-Xue Xia
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fu Fan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Liu
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiyang Yu
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jin-Chuan Hu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ben-Shang Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fei-Long Meng
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
8
|
Nguyen TTT, Tanaka Y, Sanada M, Hosaka M, Tamai M, Kagami K, Komatsu C, Somazu S, Harama D, Kasai S, Watanabe A, Akahane K, Goi K, Inukai T. CRISPR/Cas9-Mediated Induction of Relapse-Specific NT5C2 and PRPS1 Mutations Confers Thiopurine Resistance as a Relapsed Lymphoid Leukemia Model. Mol Pharmacol 2023; 103:199-210. [PMID: 36669880 DOI: 10.1124/molpharm.122.000546] [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: 04/20/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
6-Mercaptopurine (6-MP) is a key component in maintenance therapy for childhood acute lymphoblastic leukemia (ALL). Recent next-generation sequencing analysis of childhood ALL clarified the emergence of the relapse-specific mutations of the NT5C2 and PRPS1 genes, which are involved in thiopurine metabolism. In this scenario, minor clones of leukemia cells could acquire the 6-MP-resistant phenotype as a result of the NT5C2 or PRPS1 mutation during chemotherapy (including 6-MP treatment) and confer disease relapse after selective expansion. Thus, to establish new therapeutic modalities overcoming 6-MP resistance in relapsed ALL, human leukemia models with NT5C2 and PRPS1 mutations in the intrinsic genes are urgently required. Here, mimicking the initiation process of the above clinical course, we sought to induce two relapse-specific hotspot mutations (R39Q mutation of the NT5C2 gene and S103N mutation of the PRPS1 gene) into a human lymphoid leukemia cell line by homologous recombination (HR) using the CRISPR/Cas9 system. After 6-MP selection of the cells transfected with Cas9 combined with single-guide RNA and donor DNA templates specific for either of those two mutations, we obtained the sublines with the intended NT5C2-R39Q and PRPS1-S103N mutation as a result of HR. Moreover, diverse in-frame small insertion/deletions were also confirmed in the 6-MP-resistant sublines at the target sites of the NT5C2 and PRPS1 genes as a result of nonhomologous end joining. These sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations in the 6-MP sensitivity and development of therapy overcoming the thiopurine resistance of leukemia cells. SIGNIFICANCE STATEMENT: Mimicking the initiation process of relapse-specific mutations of the NT5C2 and PRPS1 genes in childhood acute lymphoblastic leukemia treated with 6-mercaptopurine (6-MP), this study sought to introduce NT5C2-R39Q and PRPS1-S103N mutations into a human lymphoid leukemia cell line by homologous recombination using the CRISPR/Cas9 system. In the resultant 6-MP-resistant sublines, the intended mutations and diverse in-frame small insertions/deletions were confirmed, indicating that the obtained sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations.
Collapse
Affiliation(s)
- Thao Thu Thi Nguyen
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Yoichi Tanaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masashi Sanada
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masumi Hosaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Minori Tamai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Keiko Kagami
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Chiaki Komatsu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shinpei Somazu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Daisuke Harama
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shin Kasai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Atsushi Watanabe
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Koushi Akahane
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Kumiko Goi
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Takeshi Inukai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| |
Collapse
|
9
|
Amodio V, Lamba S, Chilà R, Cattaneo CM, Mussolin B, Corti G, Rospo G, Berrino E, Tripodo C, Pisati F, Bartolini A, Aquilano MC, Marsoni S, Mauri G, Marchiò C, Abrignani S, Di Nicolantonio F, Germano G, Bardelli A. Genetic and pharmacological modulation of DNA mismatch repair heterogeneous tumors promotes immune surveillance. Cancer Cell 2023; 41:196-209.e5. [PMID: 36584674 PMCID: PMC9833846 DOI: 10.1016/j.ccell.2022.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/23/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022]
Abstract
Patients affected by colorectal cancer (CRC) with DNA mismatch repair deficiency (MMRd), often respond to immune checkpoint blockade therapies, while those with mismatch repair-proficient (MMRp) tumors generally do not. Interestingly, a subset of MMRp CRCs contains variable fractions of MMRd cells, but it is unknown how their presence impacts immune surveillance. We asked whether modulation of the MMRd fraction in MMR heterogeneous tumors acts as an endogenous cancer vaccine by promoting immune surveillance. To test this hypothesis, we use isogenic MMRp (Mlh1+/+) and MMRd (Mlh1-/-) mouse CRC cells. MMRp/MMRd cells mixed at different ratios are injected in immunocompetent mice and tumor rejection is observed when at least 50% of cells are MMRd. To enrich the MMRd fraction, MMRp/MMRd tumors are treated with 6-thioguanine, which leads to tumor rejection. These results suggest that genetic and pharmacological modulation of the DNA mismatch repair machinery potentiate the immunogenicity of MMR heterogeneous tumors.
Collapse
Affiliation(s)
- Vito Amodio
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Simona Lamba
- Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Rosaria Chilà
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Chiara M Cattaneo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | | | - Giorgio Corti
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Giuseppe Rospo
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy; Department of Medical Sciences, University of Torino, Torino, Italy
| | - Claudio Tripodo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139 Milan, Italy; Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90127 Palermo, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L., 20139, Milan, Italy
| | - Alice Bartolini
- Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Maria Costanza Aquilano
- Department of Hematology, Oncology, and Molecular Medicine, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Silvia Marsoni
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Gianluca Mauri
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20162 Milan, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy; Department of Medical Sciences, University of Torino, Torino, Italy
| | - Sergio Abrignani
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy
| | - Giovanni Germano
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy.
| | - Alberto Bardelli
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; Candiolo Cancer Institute, FPO - IRCCS, 10060 Candiolo, TO, Italy.
| |
Collapse
|
10
|
Gallon R, Phelps R, Betts L, Hayes C, Masic D, Irving JAE, McAnulty C, Saha V, Vora A, Wimmer K, Motwani J, Macartney C, Burn J, Jackson MS, Moorman AV, Santibanez-Koref M. Detection of constitutional mismatch repair deficiency in children and adolescents with acute lymphoblastic leukemia. Leuk Lymphoma 2023; 64:217-220. [PMID: 36272172 DOI: 10.1080/10428194.2022.2131412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Richard Gallon
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Rachel Phelps
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Leigh Betts
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Christine Hayes
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Dino Masic
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Julie A E Irving
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ciaron McAnulty
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Vaskar Saha
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Tata Translational Cancer Research Centre, Tata Medical Center, Kolkata, India
| | - Ajay Vora
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, UK
| | - Katharina Wimmer
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Jayashree Motwani
- Department of Haematology, Birmingham Women's and Children's Hospital, Birmingham, UK
| | - Christine Macartney
- Department of Paediatric Haematology and Oncology, Royal Belfast Hospital for Sick Children, Belfast, UK
| | - John Burn
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Michael S Jackson
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Anthony V Moorman
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | | |
Collapse
|
11
|
Li J, Huang F, Jiang Y, Zhao J, Wan J, Hao S. A novel costimulatory molecule gene-modified leukemia cell-derived exosome-targeted CD4 + T cell vaccine efficiently enhances anti-leukemia immunity. Front Immunol 2022; 13:1043484. [PMID: 36466863 PMCID: PMC9709463 DOI: 10.3389/fimmu.2022.1043484] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/31/2022] [Indexed: 01/06/2024] Open
Abstract
Previous studies demonstrated that CD4+ T cells can uptake tumor antigen-pulsed dendritic cell-derived exosomes (DEXO), which harbor tumor antigen peptide/pMHC I complex and costimulatory molecules and show potent effects on inducing antitumor immunity. However, in preliminary study, CD4+ T cells targeted by leukemia cell-derived exosomes (LEXs) did not show the expected effects in inducing effective anti-leukemia immunity, indicating that LEX is poorly immunogenetic largely due to an inadequate costimulatory capacity. Therefore, LEX-based anti-leukemia vaccines need to be optimized. In this study, we constructed a novel LEX-based vaccine by combining CD4+ T cells with costimulatory molecules gene-modified LEXs, which harbor upregulated CD80 and CD86, and the anti-leukemia immunity of CD80 and CD86 gene-modified LEX-targeted CD4+ T cells was investigated. We used lentiviral vectors encoding CD80 and CD86 to successfully transduced the L1210 leukemia cells, and the expression of CD80 and CD86 was remarkably upregulated in leukemia cells. The LEXs highly expressing CD80 and CD86 were obtained from the supernatants of gene-transduced leukemia cells. Our data have shown that LEX-CD8086 could promote CD4+ T cell proliferation and Th1 cytokine secretion more efficiently than control LEXs. Moreover, CD4+ TLEX-CD8086 expressed the acquired exosomal costimulatory molecules. With acquired costimulatory molecules, CD4+ TLEX-CD8086 can act as APCs and are capable of directly stimulating the leukemia cell antigen-specific CD8+ CTL response. This response was higher in potency compared to that noted by the other formulations. Furthermore, the animal study revealed that the CD4+ TLEX-CD8086 significantly inhibited tumor growth and prolonged survival of tumor-bearing mice than other formulations did in both protective and therapeutic models. In conclusion, this study revealed that CD4+ TLEX-CD8086 could effectively induce more potential anti-leukemia immunity than LEX-CD8086 alone, suggesting that the utilization of a costimulatory molecule gene-modified leukemia cell-derived exosome-targeted CD4+ T cell vaccine may have promising potential for leukemia immunotherapy.
Collapse
|
12
|
Reglero C, Dieck CL, Zask A, Forouhar F, Laurent AP, Lin WHW, Albero R, Miller HI, Ma C, Gastier-Foster JM, Loh ML, Tong L, Stockwell BR, Palomero T, Ferrando AA. Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia. Cancer Discov 2022; 12:2646-2665. [PMID: 35984649 PMCID: PMC9633388 DOI: 10.1158/2159-8290.cd-22-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/09/2022] [Accepted: 08/17/2022] [Indexed: 01/12/2023]
Abstract
Low-intensity maintenance therapy with 6-mercaptopurine (6-MP) limits the occurrence of acute lymphoblastic leukemia (ALL) relapse and is central to the success of multiagent chemotherapy protocols. Activating mutations in the 5'-nucleotidase cytosolic II (NT5C2) gene drive resistance to 6-MP in over 35% of early relapse ALL cases. Here we identify CRCD2 as a first-in-class small-molecule NT5C2 nucleotidase inhibitor broadly active against leukemias bearing highly prevalent relapse-associated mutant forms of NT5C2 in vitro and in vivo. Importantly, CRCD2 treatment also enhanced the cytotoxic activity of 6-MP in NT5C2 wild-type leukemias, leading to the identification of NT5C2 Ser502 phosphorylation as a novel NT5C2-mediated mechanism of 6-MP resistance in this disease. These results uncover an unanticipated role of nongenetic NT5C2 activation as a driver of 6-MP resistance in ALL and demonstrate the potential of NT5C2 inhibitor therapy for enhancing the efficacy of thiopurine maintenance therapy and overcoming resistance at relapse. SIGNIFICANCE Relapse-associated NT5C2 mutations directly contribute to relapse in ALL by driving resistance to chemotherapy with 6-MP. Pharmacologic inhibition of NT5C2 with CRCD2, a first-in-class nucleotidase inhibitor, enhances the cytotoxic effects of 6-MP and effectively reverses thiopurine resistance mediated by genetic and nongenetic mechanisms of NT5C2 activation in ALL. This article is highlighted in the In This Issue feature, p. 2483.
Collapse
Affiliation(s)
- Clara Reglero
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
- These authors contributed equally: Clara Reglero, Chelsea L. Dieck
| | - Chelsea L. Dieck
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
- These authors contributed equally: Clara Reglero, Chelsea L. Dieck
| | - Arie Zask
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Farhad Forouhar
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Anouchka P. Laurent
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Wen-Hsuan W. Lin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Robert Albero
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Hannah I. Miller
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Cindy Ma
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Julie M Gastier-Foster
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Children’s Oncology Group, Arcadia, CA, USA
| | - Mignon L Loh
- Division of Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapies, Seattle Children’s Hospital, University of Washington, Seattle, WA
| | - Liang Tong
- Department of Biological Sciences, Northeast Structural Genomics Consortium, Columbia University, 1212 Amsterdam Avenue, 701 Fairchild Center, New York, NY 10027, USA
| | - Brent R. Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Adolfo A. Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
13
|
How Genetics and Genomics Advances Are Rewriting Pediatric Cancer Research and Clinical Care. Medicina (B Aires) 2022; 58:medicina58101386. [PMID: 36295546 PMCID: PMC9610804 DOI: 10.3390/medicina58101386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
In the last two decades, thanks to the data that have been obtained from the Human Genome Project and the development of next-generation sequencing (NGS) technologies, research in oncology has produced extremely important results in understanding the genomic landscape of pediatric cancers, which are the main cause of death during childhood. NGS has provided significant advances in medicine by detecting germline and somatic driver variants that determine the development and progression of many types of cancers, allowing a distinction between hereditary and non-hereditary cancers, characterizing resistance mechanisms that are also related to alterations of the epigenetic apparatus, and quantifying the mutational burden of tumor cells. A combined approach of next-generation technologies allows us to investigate the numerous molecular features of the cancer cell and the effects of the environment on it, discovering and following the path of personalized therapy to defeat an "ancient" disease that has had victories and defeats. In this paper, we provide an overview of the results that have been obtained in the last decade from genomic studies that were carried out on pediatric cancer and their contribution to the more accurate and faster diagnosis in the stratification of patients and the development of new precision therapies.
Collapse
|
14
|
Ruel NM, Nguyen KH, Kim CS, Andrade LPS, Hammond JR. Impact of SLC43A3/ENBT1 Expression and Function on 6-Mercaptopurine Transport and Cytotoxicity in Human Acute Lymphoblastic Leukemia Cells. J Pharmacol Exp Ther 2022; 382:335-345. [PMID: 35798387 DOI: 10.1124/jpet.122.001155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/24/2022] [Indexed: 11/22/2022] Open
Abstract
6-Mercaptopurine (6-MP) is used extensively in the treatment of acute lymphoblastic leukemia (ALL) and inflammatory bowel diseases. Our laboratory determined previously, using a recombinant HEK293 cell model, that the SLC43A3-encoded equilibrative nucleobase transporter 1 (ENBT1) transports 6-MP into cells and significantly impacts the cytotoxicity of 6-MP in that model. To further investigate the clinical relevance of this finding, we now extend this work to an analysis of the impact of SLC43A3/ENBT1 expression and function on 6-MP uptake and cytotoxicity in leukemic lymphoblasts, the therapeutic target of 6-MP in ALL. A panel of ALL cell lines was assessed for SLC43A3/ENBT1 expression, ENBT1 function, and sensitivity to 6-MP. There was a significant difference in SLC43A3 expression among the cell lines that positively correlated with the rate of ENBT1-mediated 6-MP uptake. Cells with the lowest expression of SLC43A3 (SUP-B15: Vmax = 22± 5 pmol/µl per second) were also significantly less sensitive to 6-MP-induced cytotoxicity than were the highest expressing cells (ALL-1: Vmax = 69 ± 10 pmol/µl per second). Furthermore, knockdown of ENBT1 using short hairpin RNA interference (shRNAi) in RS4;11 cells caused a significant decrease in ENBT1-mediated 6-MP uptake (Vmax: RS4;11 = 40 ± 4 pmol/µl per second; RS4;11 shRNAi = 26 ± 3 pmol/µl per Second) and 6-MP cytotoxicity (EC50: RS4;11 = 0.58 ± 0.05 µM; RS4;11 shRNAi =1.44 ± 0.59 µM). This study showed that ENBT1 is a major contributor to 6-MP uptake in leukemia cell lines and may prove to be a biomarker for the therapeutic efficacy of 6-MP in patients with ALL. SIGNIFICANCE STATEMENT: This study shows that SLC43A3-encoded equilibrative nucleobase transporter 1 is responsible for the transport of 6-mercaptopurine (6-MP) into leukemia cells and that its level of expression can impact the cytotoxicity of 6-MP. Further studies are warranted to investigate the therapeutic implications in patient populations.
Collapse
Affiliation(s)
- Nicholas M Ruel
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Khanh Hoa Nguyen
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Chan S Kim
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Laura P S Andrade
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - James R Hammond
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Maintenance therapy for acute lymphoblastic leukemia: basic science and clinical translations. Leukemia 2022; 36:1749-1758. [PMID: 35654820 PMCID: PMC9252897 DOI: 10.1038/s41375-022-01591-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 01/21/2023]
Abstract
Maintenance therapy (MT) with oral methotrexate (MTX) and 6-mercaptopurine (6-MP) is essential for the cure of acute lymphoblastic leukemia (ALL). MTX and 6-MP interfere with nucleotide synthesis and salvage pathways. The primary cytotoxic mechanism involves the incorporation of thioguanine nucleotides (TGNs) into DNA (as DNA-TG), which may be enhanced by the inhibition of de novo purine synthesis by other MTX/6-MP metabolites. Co-medication during MT is common. Although Pneumocystis jirovecii prophylaxis appears safe, the benefit of glucocorticosteroid/vincristine pulses in improving survival and of allopurinol to moderate 6-MP pharmacokinetics remains uncertain. Numerous genetic polymorphisms influence the pharmacology, efficacy, and toxicity (mainly myelosuppression and hepatotoxicity) of MTX and thiopurines. Thiopurine S-methyltransferase (encoded by TPMT) decreases TGNs but increases methylated 6-MP metabolites (MeMPs); similarly, nudix hydrolase 15 (encoded by NUDT15) also decreases TGNs available for DNA incorporation. Loss-of-function variants in both genes are currently used to guide MT, but do not fully explain the inter-patient variability in thiopurine toxicity. Because of the large inter-individual variations in MTX/6-MP bioavailability and metabolism, dose adjustments are traditionally guided by the degree of myelosuppression, but this does not accurately reflect treatment intensity. DNA-TG is a common downstream metabolite of MTX/6-MP combination chemotherapy, and a higher level of DNA-TG has been associated with a lower relapse hazard, leading to the development of the Thiopurine Enhanced ALL Maintenance (TEAM) strategy-the addition of low-dose (2.5-12.5 mg/m2/day) 6-thioguanine to the 6-MP/MTX backbone-that is currently being tested in a randomized ALLTogether1 trial (EudraCT: 2018-001795-38). Mutations in the thiopurine and MTX metabolism pathways, and in the mismatch repair genes have been identified in early ALL relapses, providing valuable insights to assist the development of strategies to detect imminent relapse, to facilitate relapse salvage therapy, and even to bring about changes in frontline ALL therapy to mitigate this relapse risk.
Collapse
|
16
|
Toksvang LN, Als-Nielsen B, Bacon C, Bertasiute R, Duarte X, Escherich G, Helgadottir EA, Johannsdottir IR, Jónsson ÓG, Kozlowski P, Langenskjöld C, Lepik K, Niinimäki R, Overgaard UM, Punab M, Räty R, Segers H, van der Sluis I, Smith OP, Strullu M, Vaitkevičienė G, Wik HS, Heyman M, Schmiegelow K. Thiopurine Enhanced ALL Maintenance (TEAM): study protocol for a randomized study to evaluate the improvement in disease-free survival by adding very low dose 6-thioguanine to 6-mercaptopurine/methotrexate-based maintenance therapy in pediatric and adult patients (0-45 years) with newly diagnosed B-cell precursor or T-cell acute lymphoblastic leukemia treated according to the intermediate risk-high group of the ALLTogether1 protocol. BMC Cancer 2022; 22:483. [PMID: 35501736 PMCID: PMC9063225 DOI: 10.1186/s12885-022-09522-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/10/2022] [Indexed: 12/05/2022] Open
Abstract
Background A critical challenge in current acute lymphoblastic leukemia (ALL) therapy is treatment intensification in order to reduce the relapse rate in the subset of patients at the highest risk of relapse. The year-long maintenance phase is essential in relapse prevention. The Thiopurine Enhanced ALL Maintenance (TEAM) trial investigates a novel strategy for ALL maintenance. Methods TEAM is a randomized phase 3 sub-protocol to the ALLTogether1 trial, which includes patients 0–45 years of age with newly diagnosed B-cell precursor or T-cell ALL, and stratified to the intermediate risk-high (IR-high) group, in 13 European countries. In the TEAM trial, the traditional methotrexate (MTX)/6-mercaptopurine (6MP) maintenance backbone (control arm) is supplemented with low dose (2.5–12.5 mg/m2/day) oral 6-thioguanine (6TG) (experimental arm), while the starting dose of 6MP is reduced from 75 to 50 mg/m2/day. A total of 778 patients will be included in TEAM during ~ 5 years. The study will close when the last included patient has been followed for 5 years from the end of induction therapy. The primary objective of the study is to significantly improve the disease-free survival (DFS) of IR-high ALL patients by adding 6TG to 6MP/MTX-based maintenance therapy. TEAM has 80% power to detect a 7% increase in 5-year DFS through a 50% reduction in relapse rate. DFS will be evaluated by intention-to-treat analysis. In addition to reducing relapse, TEAM may also reduce hepatotoxicity and hypoglycemia caused by high levels of methylated 6MP metabolites. Methotrexate/6MP metabolites will be monitored and low levels will be reported back to clinicians to identify potentially non-adherent patients. Discussion TEAM provides a novel strategy for maintenance therapy in ALL with the potential of improving DFS through reducing relapse rate. Potential risk factors that have been considered include hepatic sinusoidal obstruction syndrome/nodular regenerative hyperplasia, second cancer, infection, and osteonecrosis. Metabolite monitoring can potentially increase treatment adherence in both treatment arms. Trial registration EudraCT, 2018–001795-38. Registered 2020-05-15, Clinicaltrials.gov, NCT04307576. Registered 2020-03-13, https://clinicaltrials.gov/ct2/show/NCT04307576 Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09522-3.
Collapse
Affiliation(s)
- Linea Natalie Toksvang
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | | | - Ruta Bertasiute
- Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Ximo Duarte
- Instituto Português de Oncologia Lisboa Francisco Gentil Departamento de Pediatria, Lisbon, Portugal
| | | | | | | | | | | | | | | | - Riitta Niinimäki
- Oulu University Hospital and PEDEGRO Research Unit, University of Oulu, Oulu, Finland
| | | | - Mari Punab
- Tartu University Hospital, Tartu, Estonia
| | - Riikka Räty
- Helsinki University Central Hospital, Helsinki, Finland
| | - Heidi Segers
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | | | | | - Marion Strullu
- Université de Paris, hôpital universitaire Robert-Debré (APHP), Paris, France
| | - Goda Vaitkevičienė
- Center for Pediatric Oncology and Hematology, Vilnius University, Vilnius, Lithuania
| | | | - Mats Heyman
- Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.,University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Mas-Ponte D, McCullough M, Supek F. Spectrum of DNA mismatch repair failures viewed through the lens of cancer genomics and implications for therapy. Clin Sci (Lond) 2022; 136:383-404. [PMID: 35274136 PMCID: PMC8919091 DOI: 10.1042/cs20210682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
Genome sequencing can be used to detect DNA repair failures in tumors and learn about underlying mechanisms. Here, we synthesize findings from genomic studies that examined deficiencies of the DNA mismatch repair (MMR) pathway. The impairment of MMR results in genome-wide hypermutation and in the 'microsatellite instability' (MSI) phenotype-occurrence of indel mutations at short tandem repeat (microsatellite) loci. The MSI status of tumors was traditionally assessed by molecular testing of a selected set of MS loci or by measuring MMR protein expression levels. Today, genomic data can provide a more complete picture of the consequences on genomic instability. Multiple computational studies examined somatic mutation distributions that result from failed DNA repair pathways in tumors. These include analyzing the commonly studied trinucleotide mutational spectra of single-nucleotide variants (SNVs), as well as of other features such as indels, structural variants, mutation clusters and regional mutation rate redistribution. The identified mutation patterns can be used to rigorously measure prevalence of MMR failures across cancer types, and potentially to subcategorize the MMR deficiencies. Diverse data sources, genomic and pre-genomic, from human and from experimental models, suggest there are different ways in which MMR can fail, and/or that the cell-type or genetic background may result in different types of MMR mutational patterns. The spectrum of MMR failures may direct cancer evolution, generating particular sets of driver mutations. Moreover, MMR affects outcomes of therapy by DNA damaging drugs, antimetabolites, nonsense-mediated mRNA decay (NMD) inhibitors, and immunotherapy by promoting either resistance or sensitivity, depending on the type of therapy.
Collapse
Affiliation(s)
- David Mas-Ponte
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Marcel McCullough
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Fran Supek
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Pg Lluís Companys, 23, Barcelona 08010, Spain
| |
Collapse
|
18
|
Resistance Mechanisms in Pediatric B-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2022; 23:ijms23063067. [PMID: 35328487 PMCID: PMC8950780 DOI: 10.3390/ijms23063067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the rapid development of medicine, even nowadays, acute lymphoblastic leukemia (ALL) is still a problem for pediatric clinicians. Modern medicine has reached a limit of curability even though the recovery rate exceeds 90%. Relapse occurs in around 20% of treated patients and, regrettably, 10% of diagnosed ALL patients are still incurable. In this article, we would like to focus on the treatment resistance and disease relapse of patients with B-cell leukemia in the context of prognostic factors of ALL. We demonstrate the mechanisms of the resistance to steroid therapy and Tyrosine Kinase Inhibitors and assess the impact of genetic factors on the treatment resistance, especially TCF3::HLF translocation. We compare therapeutic protocols and decipher how cancer cells become resistant to innovative treatments—including CAR-T-cell therapies and monoclonal antibodies. The comparisons made in our article help to bring closer the main factors of resistance in hematologic malignancies in the context of ALL.
Collapse
|
19
|
Kroeze E, Weijers DD, Hagleitner MM, de Groot-Kruseman HA, Jongmans MCJ, Kuiper RP, Pieters R, Meijerink JPP, Loeffen JLC. High Prevalence of Constitutional Mismatch Repair Deficiency in a Pediatric T-cell Lymphoblastic Lymphoma Cohort. Hemasphere 2021; 6:e668. [PMID: 34964038 PMCID: PMC8697338 DOI: 10.1097/hs9.0000000000000668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022] Open
Abstract
This study describes the clinical characteristics of a complete Dutch T-cell lymphoblastic lymphoma (T-LBL) cohort, including second primary malignancies and comorbidities. We show that over 10% of patients in this complete T-LBL cohort have been diagnosed with a cancer predisposition syndrome (CPS), consisting almost exclusively of constitutional mismatch repair deficiency (CMMRD). The clinical characteristics of sporadic T-LBL patients were compared with T-LBL patients that have been diagnosed with CMMRD. This shows that disease presentation is comparable but that disease localization in CMMRD patients might be more localized. The percentage of CPS seems reliable considering the completeness of the cohort of Dutch T-LBL patients and might even be an underestimation (possibility of undiagnosed CPS patients in cohort). As the frequency of an underlying predisposition syndrome among T-LBL patients may be underestimated at present, we advocate for screening all pediatric T-LBL patients for the presence of germline mutations in mismatch repair genes.
Collapse
Affiliation(s)
- Emma Kroeze
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Dilys D Weijers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | - Marjolijn C J Jongmans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Roland P Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Jan L C Loeffen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
20
|
Skhoun H, Khattab M, Belkhayat A, Takki Chebihi Z, Dakka N, El Baghdadi J. A prognostic approach on a case of pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia with monosomy-7. Clin Case Rep 2021; 9:e05207. [PMID: 34963805 PMCID: PMC8710846 DOI: 10.1002/ccr3.5207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/04/2021] [Accepted: 11/14/2021] [Indexed: 11/21/2022] Open
Abstract
In this work, we present the first case of a Ph-positive ALL Moroccan girl with t(9;22)(q34;q11) and monosomy-7. She was diagnosed with Ph-positive ALL based on bone marrow examination, immunophenotyping, and cytogenetic analysis. She relapsed after treatment with the persistence of the Ph chromosome and the appearance of a monosomy-7.
Collapse
Affiliation(s)
- Hanaa Skhoun
- Genetics UnitMilitary Hospital Mohammed VRabatMorocco
- Laboratory of Human Pathologies BiologyGenomic Center of Human PathologiesFaculty of SciencesMohammed V University in RabatRabatMorocco
| | - Mohammed Khattab
- Department of Pediatric Hemato‐OncologyChildren's Hospital of RabatRabatMorocco
| | | | | | - Nadia Dakka
- Laboratory of Human Pathologies BiologyGenomic Center of Human PathologiesFaculty of SciencesMohammed V University in RabatRabatMorocco
| | | |
Collapse
|
21
|
DFF40 deficiency in cancerous T cells is implicated in chemotherapy drug sensitivity and resistance through the regulation of the apoptotic pathway. Biochem Pharmacol 2021; 194:114801. [PMID: 34678222 DOI: 10.1016/j.bcp.2021.114801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
The regulation of the apoptotic pathway is one of the most studied mechanisms regarding cancer cell resistance. Many mutations have been linked to drug resistance. The DNA fragmentation factor 40 (DFF40) has been gaining interest regarding cancer cell response to chemotherapy and patient outcomes. Glioblastomas and uterine leiomyosarcomas have been shown to have a downregulation of DFF40 expression, conferring a poor patient prognosis. In concordance with these observations, in this study, we showed that DFF40 gene is also downregulated in breast, endocervical, ovarian, lung, pancreas and glioblastomas. DFF40 is the endonuclease responsible of DNA fragmentation during apoptosis. In this study, we sought to determine if a DFF40 deficiency in Jurkat T cells could impact the sensitivity to conventional chemotherapy drugs. CRISPR-cas9 generated DFF40 knockout (DFF40 KO) stable Jurkat cells and wild-type (DFF40 WT) cells were treated with different antimetabolites and topoisomerase II (TOP2) inhibitors, and cell viability was subsequently assessed. DFF40 deficient cells show chemoresistance to antimetabolites (e.g. methotrexate, 6-mercaptopurine and cytarabine) and surprisingly, they are more sensitive to TOP2 inhibitors (e.g. etoposide and teniposide). DFF40 deficient cells exposed to cytarabine present lower phosphatidylserine translocation levels to the outer cell membrane layer. Etoposide exposure in DFF40 deficient cells induces higher mortality levels and downregulation of Bcl-xL cells compared to DFF40 expressing T cells. The abolition of DFF40 expression in Jurkat cells significantly impairs histone H2AX phosphorylation following etoposide and cytarabine treatments. Our findings suggest that DFF40 is a novel key target in cancer cell resistance that potentially regulates genomic stability.
Collapse
|
22
|
Affiliation(s)
- Nikki A Evensen
- Perlmutter Cancer Center, NYU Grossman School of Medicine New York, New York, NY, USA
| | - William L Carroll
- Perlmutter Cancer Center, NYU Grossman School of Medicine New York, New York, NY, USA.
- Department of Pediatrics and Pathology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
23
|
Yang F, Brady SW, Tang C, Sun H, Du L, Barz MJ, Ma X, Chen Y, Fang H, Li X, Kolekar P, Pathak O, Cai J, Ding L, Wang T, von Stackelberg A, Shen S, Eckert C, Klco JM, Chen H, Duan C, Liu Y, Li H, Li B, Kirschner-Schwabe R, Zhang J, Zhou BBS. Chemotherapy and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse. NATURE CANCER 2021; 2:819-834. [PMID: 35122027 DOI: 10.1038/s43018-021-00230-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/02/2021] [Indexed: 06/14/2023]
Abstract
Chemotherapy is a standard treatment for pediatric acute lymphoblastic leukemia (ALL), which sometimes relapses with chemoresistant features. However, whether acquired drug-resistance mutations in relapsed ALL pre-exist or are induced by treatment remains unknown. Here we provide direct evidence of a specific mechanism by which chemotherapy induces drug-resistance-associated mutations leading to relapse. Using genomic and functional analysis of relapsed ALL we show that thiopurine treatment in mismatch repair (MMR)-deficient leukemias induces hotspot TP53 R248Q mutations through a specific mutational signature (thio-dMMR). Clonal evolution analysis reveals sequential MMR inactivation followed by TP53 mutation in some patients with ALL. Acquired TP53 R248Q mutations are associated with on-treatment relapse, poor treatment response and resistance to multiple chemotherapeutic agents, which could be reversed by pharmacological p53 reactivation. Our findings indicate that TP53 R248Q in relapsed ALL originates through synergistic mutagenesis from thiopurine treatment and MMR deficiency and suggest strategies to prevent or treat TP53-mutant relapse.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Samuel W Brady
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chao Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiying Sun
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijuan Du
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Malwine J Barz
- Department of Pediatric Oncology/Hematology, Charite-Universitaetsmedizin Berlin, Berlin, Germany
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yao Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomeng Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pandurang Kolekar
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Omkar Pathak
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiaoyang Cai
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixia Ding
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Wang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Arend von Stackelberg
- Department of Pediatric Oncology/Hematology, Charite-Universitaetsmedizin Berlin, Berlin, Germany
| | - Shuhong Shen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cornelia Eckert
- Department of Pediatric Oncology/Hematology, Charite-Universitaetsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Caiwen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Liu
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Benshang Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renate Kirschner-Schwabe
- Department of Pediatric Oncology/Hematology, Charite-Universitaetsmedizin Berlin, Berlin, Germany.
- German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany.
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
24
|
Oshima K, Zhao J, Pérez-Durán P, Brown JA, Patiño-Galindo JA, Chu T, Quinn A, Gunning T, Belver L, Ambesi-Impiombato A, Tosello V, Wang Z, Sulis ML, Kato M, Koh K, Paganin M, Basso G, Balbin M, Nicolas C, Gastier-Foster JM, Devidas M, Loh ML, Paietta E, Tallman MS, Rowe JM, Litzow M, Minden MD, Meijerink J, Rabadan R, Ferrando A. Mutational and functional genetics mapping of chemotherapy resistance mechanisms in relapsed acute lymphoblastic leukemia. NATURE CANCER 2020; 1:1113-1127. [PMID: 33796864 PMCID: PMC8011577 DOI: 10.1038/s43018-020-00124-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
Multi-agent combination chemotherapy can be curative in acute lymphoblastic leukemia (ALL). Still, patients with primary refractory disease or with relapsed leukemia have a very poor prognosis. Here we integrate an in-depth dissection of the mutational landscape across diagnostic and relapsed pediatric and adult ALL samples with genome-wide CRISPR screen analysis of gene-drug interactions across seven ALL chemotherapy drugs. By combining these analyses, we uncover diagnostic and relapse-specific mutational mechanisms as well as genetic drivers of chemoresistance. Functionally, our data identifies common and drug-specific pathways modulating chemotherapy response and underscores the effect of drug combinations in restricting the selection of resistance-driving genetic lesions. In addition, by identifying actionable targets for the reversal of chemotherapy resistance, these analyses open novel therapeutic opportunities for the treatment of relapse and refractory disease.
Collapse
Affiliation(s)
- Koichi Oshima
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Pablo Pérez-Durán
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Jessie A Brown
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Juan Angel Patiño-Galindo
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Timothy Chu
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Aidan Quinn
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Thomas Gunning
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Laura Belver
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | | | | | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Maria Luisa Sulis
- Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Motohiro Kato
- Department of Hematology-Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Katsuyoshi Koh
- Department of Hematology-Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Maddalena Paganin
- Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padova, Italy
- Haematology-Oncology Division, Department of Woman's and Child's Health, University Hospital of Padua, Padua, Italy
| | - Giuseppe Basso
- Haematology-Oncology Division, Department of Woman's and Child's Health, University Hospital of Padua, Padua, Italy
- IIGM Italian Institute of Genomic Medicine, Turin, Italy
| | - Milagros Balbin
- Molecular Oncology Laboratory, Instituto Universitario de Oncologia del Principado de Asturias, Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Concepcion Nicolas
- Hematology Service, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Julie M Gastier-Foster
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Departments of Pathology and Pediatrics, Ohio State University School of Medicine, Columbus, OH, USA
- Children's Oncology Group, Arcadia, CA, USA
| | - Meenakshi Devidas
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mignon L Loh
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - Martin S Tallman
- Department of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Mark D Minden
- Department of Oncology/Hematology, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Jules Meijerink
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Department of Pediatrics, Columbia University, New York, NY, USA.
| |
Collapse
|
25
|
Jasinski S, De Los Reyes FA, Yametti GC, Pierro J, Raetz E, Carroll WL. Immunotherapy in Pediatric B-Cell Acute Lymphoblastic Leukemia: Advances and Ongoing Challenges. Paediatr Drugs 2020; 22:485-499. [PMID: 32860590 PMCID: PMC7537790 DOI: 10.1007/s40272-020-00413-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.
Collapse
Affiliation(s)
- Sylwia Jasinski
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | | | - Gloria Contreras Yametti
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Joanna Pierro
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - William L Carroll
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
26
|
Rahimian E, Amini A, Alikarami F, Pezeshki SMS, Saki N, Safa M. DNA repair pathways as guardians of the genome: Therapeutic potential and possible prognostic role in hematologic neoplasms. DNA Repair (Amst) 2020; 96:102951. [PMID: 32971475 DOI: 10.1016/j.dnarep.2020.102951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022]
Abstract
DNA repair pathways, which are also identified as guardians of the genome, protect cells from frequent damage that can lead to DNA breaks. The most deleterious types of damage are double-strand breaks (DSBs), which are repaired by homologous recombination (HR) and non-homologous end joining (NHEJ). Single strand breaks (SSBs) can be corrected through base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR). Failure to restore DNA lesions or inappropriately repaired DNA damage culminates in genomic instability and changes in the regulation of cellular functions. Intriguingly, particular mutations and translocations are accompanied by special types of leukemia. Besides, expression patterns of certain repair genes are altered in different hematologic malignancies. Moreover, analysis of mutations in key mediators of DNA damage repair (DDR) pathways, as well as investigation of their expression and function, may provide us with emerging biomarkers of response/resistance to treatment. Therefore, defective DDR pathways can offer a rational starting point for developing DNA repair-targeted drugs. In this review, we address genetic alterations and gene/protein expression changes, as well as provide an overview of DNA repair pathways.
Collapse
Affiliation(s)
- Elahe Rahimian
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Amini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Alikarami
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA 19104, USA
| | - Seyed Mohammad Sadegh Pezeshki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Waanders E, Gu Z, Dobson SM, Antić Ž, Crawford JC, Ma X, Edmonson MN, Payne-Turner D, van de Vorst M, Jongmans MCJ, McGuire I, Zhou X, Wang J, Shi L, Pounds S, Pei D, Cheng C, Song G, Fan Y, Shao Y, Rusch M, McCastlain K, Yu J, van Boxtel R, Blokzijl F, Iacobucci I, Roberts KG, Wen J, Wu G, Ma J, Easton J, Neale G, Olsen SR, Nichols KE, Pui CH, Zhang J, Evans WE, Relling MV, Yang JJ, Thomas PG, Dick JE, Kuiper RP, Mullighan CG. Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia. Blood Cancer Discov 2020; 1:96-111. [PMID: 32793890 PMCID: PMC7418874 DOI: 10.1158/0008-5472.bcd-19-0041] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/22/2019] [Accepted: 11/08/2019] [Indexed: 01/25/2023] Open
Abstract
Relapse of acute lymphoblastic leukemia (ALL) remains a leading cause of childhood death. Prior studies have shown clonal mutations at relapse often arise from relapse-fated subclones that exist at diagnosis. However, the genomic landscape, evolutionary trajectories and mutational mechanisms driving relapse are incompletely understood. In an analysis of 92 cases of relapsed childhood ALL, incorporating multimodal DNA and RNA sequencing, deep digital mutational tracking and xenografting to formally define clonal structure, we identify 50 significant targets of mutation with distinct patterns of mutational acquisition or enrichment. CREBBP, NOTCH1, and Ras signaling mutations rose from diagnosis subclones, whereas variants in NCOR2, USH2A and NT5C2 were exclusively observed at relapse. Evolutionary modeling and xenografting demonstrated that relapse-fated clones were minor (50%), major (27%) or multiclonal (18%) at diagnosis. Putative second leukemias, including those with lineage shift, were shown to most commonly represent relapse from an ancestral clone rather than a truly independent second primary leukemia. A subset of leukemias prone to repeated relapse exhibited hypermutation driven by at least three distinct mutational processes, resulting in heightened neoepitope burden and potential vulnerability to immunotherapy. Finally, relapse-driving sequence mutations were detected prior to relapse using deep digital PCR at levels comparable to orthogonal approaches to monitor levels of measurable residual disease. These results provide a genomic framework to anticipate and circumvent relapse by earlier detection and targeting of relapse-fated clones.
Collapse
Affiliation(s)
- Esmé Waanders
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Zhaohui Gu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stephanie M Dobson
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Željko Antić
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael N Edmonson
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Debbie Payne-Turner
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Maartje van de Vorst
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Marjolijn C J Jongmans
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Irina McGuire
- Department of Information Services, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jian Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Guangchun Song
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ying Shao
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kelly McCastlain
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jiangyan Yu
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Francis Blokzijl
- Oncode Institute, University Medical Center Utrecht, Utrecht, the Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ji Wen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gang Wu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Geoffrey Neale
- The Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott R Olsen
- The Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - William E Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Roland P Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
28
|
Abstract
Genomic analyses have revolutionized our understanding of the biology of B-progenitor acute lymphoblastic leukemia (ALL). Studies of thousands of cases across the age spectrum have revised the taxonomy of B-ALL by identifying multiple new subgroups with diverse sequence and structural initiating events that vary substantially by age at diagnosis and prognostic significance. There is a growing appreciation of the role of inherited genetic variation in predisposition to ALL and drug responsiveness and of the nature of genetic variegation and clonal evolution that may be targeted for improved diagnostic, risk stratification, disease monitoring, and therapeutic intervention. This review provides an overview of the current state of knowledge of the genetic basis of B-ALL, with an emphasis on recent discoveries that have changed our approach to diagnosis and monitoring.
Collapse
Affiliation(s)
- Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
29
|
Witkowski MT, Dolgalev I, Evensen NA, Ma C, Chambers T, Roberts KG, Sreeram S, Dai Y, Tikhonova AN, Lasry A, Qu C, Pei D, Cheng C, Robbins GA, Pierro J, Selvaraj S, Mezzano V, Daves M, Lupo PJ, Scheurer ME, Loomis CA, Mullighan CG, Chen W, Rabin KR, Tsirigos A, Carroll WL, Aifantis I. Extensive Remodeling of the Immune Microenvironment in B Cell Acute Lymphoblastic Leukemia. Cancer Cell 2020; 37:867-882.e12. [PMID: 32470390 PMCID: PMC7341535 DOI: 10.1016/j.ccell.2020.04.015] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/05/2020] [Accepted: 04/22/2020] [Indexed: 12/23/2022]
Abstract
A subset of B cell acute lymphoblastic leukemia (B-ALL) patients will relapse and succumb to therapy-resistant disease. The bone marrow microenvironment may support B-ALL progression and treatment evasion. Utilizing single-cell approaches, we demonstrate B-ALL bone marrow immune microenvironment remodeling upon disease initiation and subsequent re-emergence during conventional chemotherapy. We uncover a role for non-classical monocytes in B-ALL survival, and demonstrate monocyte abundance at B-ALL diagnosis is predictive of pediatric and adult B-ALL patient survival. We show that human B-ALL blasts alter a vascularized microenvironment promoting monocytic differentiation, while depleting leukemia-associated monocytes in B-ALL animal models prolongs disease remission in vivo. Our profiling of the B-ALL immune microenvironment identifies extrinsic regulators of B-ALL survival supporting new immune-based therapeutic approaches for high-risk B-ALL treatment.
Collapse
Affiliation(s)
- Matthew T Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| | - Igor Dolgalev
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY 10016, USA
| | - Nikki A Evensen
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY 11202, USA; Department of Biomedical Engineering, New York University, New York, NY 11202, USA
| | - Tiffany Chambers
- Division of Pediatric Hematology/Oncology, College of Medicine, Baylor University, Houston, TX 77030, USA
| | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sheetal Sreeram
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Yuling Dai
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Anastasia N Tikhonova
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Audrey Lasry
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Chunxu Qu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Deqing Pei
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cheng Cheng
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Gabriel A Robbins
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Joanna Pierro
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Shanmugapriya Selvaraj
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Experimental Pathology Research Laboratory, New York University School of Medicine, New York, NY 10016, USA
| | - Valeria Mezzano
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Experimental Pathology Research Laboratory, New York University School of Medicine, New York, NY 10016, USA
| | - Marla Daves
- Division of Pediatric Hematology/Oncology, College of Medicine, Baylor University, Houston, TX 77030, USA
| | - Philip J Lupo
- Division of Pediatric Hematology/Oncology, College of Medicine, Baylor University, Houston, TX 77030, USA
| | - Michael E Scheurer
- Division of Pediatric Hematology/Oncology, College of Medicine, Baylor University, Houston, TX 77030, USA
| | - Cynthia A Loomis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Experimental Pathology Research Laboratory, New York University School of Medicine, New York, NY 10016, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY 11202, USA; Department of Biomedical Engineering, New York University, New York, NY 11202, USA
| | - Karen R Rabin
- Division of Pediatric Hematology/Oncology, College of Medicine, Baylor University, Houston, TX 77030, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, New York University School of Medicine, New York, NY 10016, USA
| | - William L Carroll
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
30
|
Pierro J, Saliba J, Narang S, Sethia G, Saint Fleur-Lominy S, Chowdhury A, Qualls A, Fay H, Kilberg HL, Moriyama T, Fuller TJ, Teachey DT, Schmiegelow K, Yang JJ, Loh ML, Brown PA, Zhang J, Ma X, Tsirigos A, Evensen NA, Carroll WL. The NSD2 p.E1099K Mutation Is Enriched at Relapse and Confers Drug Resistance in a Cell Context-Dependent Manner in Pediatric Acute Lymphoblastic Leukemia. Mol Cancer Res 2020; 18:1153-1165. [PMID: 32332049 DOI: 10.1158/1541-7786.mcr-20-0092] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/10/2020] [Accepted: 04/17/2020] [Indexed: 11/16/2022]
Abstract
The NSD2 p.E1099K (EK) mutation is observed in 10% of acute lymphoblastic leukemia (ALL) samples with enrichment at relapse indicating a role in clonal evolution and drug resistance. To discover mechanisms that mediate clonal expansion, we engineered B-precursor ALL (B-ALL) cell lines (Reh, 697) to overexpress wildtype (WT) and EK NSD2, but observed no differences in proliferation, clonal growth, or chemosensitivity. To address whether NSD2 EK acts collaboratively with other pathways, we used short hairpin RNAs to knockdown expression of NSD2 in B-ALL cell lines heterozygous for NSD2 EK (RS4;11, RCH-ACV, SEM). Knockdown resulted in decreased proliferation in all lines, decreased clonal growth in RCH-ACV, and increased sensitivity to cytotoxic chemotherapeutic agents, although the pattern of drug sensitivity varied among cell lines implying that the oncogenic properties of NSD2 mutations are likely cell context specific and rely on cooperative pathways. Knockdown of both Type II and REIIBP EK isoforms had a greater impact than knockdown of Type II alone, suggesting that both SET containing EK isoforms contribute to phenotypic changes driving relapse. Furthermore, in vivo models using both cell lines and patient samples revealed dramatically enhanced proliferation of NSD2 EK compared with WT and reduced sensitivity to 6-mercaptopurine in the relapse sample relative to diagnosis. Finally, EK-mediated changes in chromatin state and transcriptional output differed dramatically among cell lines further supporting a cell context-specific role of NSD2 EK. These results demonstrate a unique role of NSD2 EK in mediating clonal fitness through pleiotropic mechanisms dependent on the genetic and epigenetic landscape. IMPLICATIONS: NSD2 EK mutation leads to drug resistance and a clonal advantage in childhood B-ALL.
Collapse
Affiliation(s)
- Joanna Pierro
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York.,Division of Pediatric Hematology/Oncology, Hassenfeld Children's Hospital at NYU Langone Health, New York, New York
| | - Jason Saliba
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Sonali Narang
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Gunjan Sethia
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Shella Saint Fleur-Lominy
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York.,Division of Medical Hematology/Oncology, NYU Langone Health, New York, New York
| | - Ashfiyah Chowdhury
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Anita Qualls
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Hannah Fay
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Harrison L Kilberg
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Takaya Moriyama
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Tori J Fuller
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania
| | - David T Teachey
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital and The Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California
| | - Patrick A Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jinghui Zhang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xiaotu Ma
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Aristotelis Tsirigos
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Nikki A Evensen
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - William L Carroll
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York. .,Division of Pediatric Hematology/Oncology, Hassenfeld Children's Hospital at NYU Langone Health, New York, New York
| |
Collapse
|
31
|
Witkowski MT, Lasry A, Carroll WL, Aifantis I. Immune-Based Therapies in Acute Leukemia. Trends Cancer 2019; 5:604-618. [PMID: 31706508 PMCID: PMC6859901 DOI: 10.1016/j.trecan.2019.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
Treatment resistance remains a leading cause of acute leukemia-related deaths. Thus, there is an unmet need to develop novel approaches to improve outcome. New immune-based therapies with chimeric antigen receptor (CAR) T cells, bi-specific T cell engagers (BiTEs), and immune checkpoint blockers (ICBs) have emerged as effective treatment options for chemoresistant B cell acute lymphoblastic leukemia (B-ALL) and acute myeloid leukemia (AML). However, many patients show resistance to these immune-based approaches. This review describes crucial lessons learned from immune-based approaches targeting high-risk B-ALL and AML, such as the leukemia-intrinsic (e.g., target antigen loss, tumor heterogeneity) and -extrinsic (e.g., immunosuppressive microenvironment) mechanisms that drive treatment resistance, and discusses alternative approaches to enhance the effectiveness of these immune-based treatment regimens.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Disease Susceptibility
- Humans
- Immunity
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Molecular Targeted Therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Matthew T Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| | - Audrey Lasry
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - William L Carroll
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Department of Pediatrics, New York University School of Medicine, New York, NY 10016, USA
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
32
|
Zhang Y, Li X, Bai L, Li L, Li D, Ding X, Wang B, Li C. MicroRNA-31 is a potential biomarker for screening B-lymphoblastic leukemia in children. Oncol Lett 2019; 18:4930-4935. [PMID: 31612004 DOI: 10.3892/ol.2019.10843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/11/2019] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to investigate the expression and significance of microRNA-31 (miR-31) in children with acute B-lymphoblastic leukemia (B-ALL). Bone marrow specimens and peripheral blood were collected from children with B-ALL (n=38) and healthy controls (n=18). Total RNA was extracted and the expression levels of miR-31 were measured using quantitative PCR. In addition, a receiver operating characteristic curve was generated, and the area under the curve (AUC) was calculated to evaluate the diagnostic value of miR-31 for the development of B-ALL. miR-31 expression was significantly lower in the B-ALL group compared with in the control group (P<0.05). Additionally, the expression levels of miR-31 in the B-ALL group before treatment were markedly lower than in the B-ALL group after treatment, and miR-31 expression was significantly lower after 30 days of treatment compared with after 12 weeks of treatment. Furthermore, miR-31 expression in the group of children ≥10 years of age was higher than that in the group of children <10 years of age. Furthermore, the expression levels of miR-31 were higher in the low-risk group compared with in the medium- and high-risk groups (P<0.05). When the cutoff value was set at 1.8, the AUC of miR-31 for B-ALL diagnosis was 0.915 (95% CI, 0.828-1.000; P<0.0001), with a sensitivity and specificity of 80.8 and 100%, respectively. In conclusion, miR-31 may exert an anticancer role in B-ALL in children and may be a potential marker to assist in diagnosis and prognostic prediction.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xin Li
- Department of Pediatric Medicine, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Liping Bai
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Li Li
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Danhong Li
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xue Ding
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Bo Wang
- Department of Hematology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Chengwei Li
- Department of Clinical Laboratory, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
33
|
Genetics and mechanisms of NT5C2-driven chemotherapy resistance in relapsed ALL. Blood 2019; 133:2263-2268. [PMID: 30910786 DOI: 10.1182/blood-2019-01-852392] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/18/2019] [Indexed: 01/01/2023] Open
Abstract
Mutations in the cytosolic 5' nucleotidase II (NT5C2) gene drive resistance to thiopurine chemotherapy in relapsed acute lymphoblastic leukemia (ALL). Mechanistically, NT5C2 mutant proteins have increased nucleotidase activity as a result of altered activating and autoregulatory switch-off mechanisms. Leukemias with NT5C2 mutations are chemoresistant to 6-mercaptopurine yet show impaired proliferation and self-renewal. Direct targeting of NT5C2 or inhibition of compensatory pathways active in NT5C2 mutant cells may antagonize the emergence of NT5C2 mutant clones driving resistance and relapse in ALL.
Collapse
|
34
|
Mallory N, Pierro J, Raetz E, Carroll WL. The potential of precision medicine for childhood acute lymphoblastic leukemia: opportunities and challenges. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1547108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Nicole Mallory
- Perlmutter Cancer Center and Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| | - Joanna Pierro
- Perlmutter Cancer Center and Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center and Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| | - William L. Carroll
- Perlmutter Cancer Center and Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, New York, NY, USA
| |
Collapse
|