193 results on '"Weinstock DM"'
Search Results
2. Induction of chromosomal translocations in mouse and human cells using site-specific endonucleases.
- Author
-
Weinstock DM, Brunet E, and Jasin M
- Published
- 2008
3. Prolonged shedding of multidrug-resistant influenza A virus in an immunocompromised patient.
- Author
-
Weinstock DM, Gubareva LV, and Zuccotti G
- Published
- 2003
4. Asciminib plus dasatinib and prednisone for Philadelphia chromosome-positive acute leukemia.
- Author
-
Luskin MR, Murakami MA, Keating J, Flamand Y, Winer ES, Garcia JS, Stahl M, Stone RM, Wadleigh M, Jaeckle SL, Hagopian E, Weinstock DM, Liegel J, McMasters M, Wang ES, Stock W, and DeAngelo DJ
- Subjects
- Humans, Middle Aged, Female, Aged, Male, Adult, Aged, 80 and over, Philadelphia Chromosome, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl antagonists & inhibitors, Niacinamide analogs & derivatives, Pyrazoles, Dasatinib administration & dosage, Dasatinib adverse effects, Dasatinib therapeutic use, Prednisone administration & dosage, Prednisone adverse effects, Prednisone therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Abstract: Dasatinib is an effective treatment for Philadelphia chromosome-positive (Ph+) acute leukemia, but some patients develop resistance. Combination treatment with dasatinib and asciminib, an allosteric inhibitor of BCR::ABL1, may deepen responses and prevent the emergence of dasatinib-resistant clones. In this phase 1 study (NCT03595017), 24 adults with Ph+ acute lymphoblastic leukemia (ALL; n = 22; p190, n = 16; p210, n = 6) and chronic myeloid leukemia in lymphoid blast crisis (n = 2) were treated with escalating daily doses of asciminib in combination with dasatinib 140 mg daily plus prednisone 60 mg/m2 daily to determine the maximum tolerated dose. After a 28-day induction, dasatinib and asciminib were continued indefinitely or until hematopoietic stem cell transplant. The median age was 64.5 years (range, 33-85; 50% aged ≥65 years). The recommended phase 2 dose of asciminib was 80 mg daily in combination with dasatinib and prednisone. The dose limiting toxicity at 160 mg daily was asymptomatic grade 3 pancreatic enzyme elevation without symptomatic pancreatitis. There were no vaso-occlusive events. Among patients with de novo ALL, the complete hematologic remission rates at days 28 and 84 were 84% and 100%, respectively. At day 84, 100% of patients achieved complete cytogenetic remission, 89% achieved measurable residual disease negativity (<0.01%) by multicolor flow cytometry, and 74% and 26% achieved BCR::ABL1 reverse transcription quantitative polymerase chain reaction <0.1% and <0.01%, respectively. Dual BCR::ABL1 inhibition with dasatinib and asciminib is safe with encouraging activity in patients with de novo Ph+ ALL. This trial was registered at www.clinicaltrials.gov as #NCT02081378., (© 2025 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2025
- Full Text
- View/download PDF
5. Author Correction: Duvelisib plus romidepsin in relapsed/refractory T cell lymphomas: a phase 1b/2a trial.
- Author
-
Horwitz SM, Nirmal AJ, Rahman J, Xu R, Drill E, Galasso N, Ganesan N, Davey T, Hancock H, Perez L, Maccaro C, Bahgat A, Marzouk E, Cathcart E, Moskowitz A, Noy A, Kumar A, Jacobsen E, Fisher DC, Mehta-Shah N, Kim YH, Khodadoust M, Kotlov N, Nikitina A, Kudryashova O, Zubareva V, Zornikova K, Shin N, Sorokina M, Degryse S, Postovalova E, Bagaev A, Hosszu K, McAvoy D, Boelens JJ, Wu W, Ciantra Z, Appelt JW, Trevisani C, Amaka S, Weinstock DM, and Vardhana SA
- Published
- 2024
- Full Text
- View/download PDF
6. Duvelisib plus romidepsin in relapsed/refractory T cell lymphomas: a phase 1b/2a trial.
- Author
-
Horwitz SM, Nirmal AJ, Rahman J, Xu R, Drill E, Galasso N, Ganesan N, Davey T, Hancock H, Perez L, Maccaro C, Bahgat A, Marzouk E, Cathcart E, Moskowitz A, Noy A, Kumar A, Jacobsen E, Fisher DC, Mehta-Shah N, Kim YH, Khodadoust M, Kotlov N, Nikitina A, Kudryashova O, Zubareva V, Zornikova K, Shin N, Sorokina M, Degryse S, Postovalova E, Bagaev A, Hosszu K, McAvoy D, Boelens JJ, Wu W, Ciantra Z, Appelt JW, Trevisani C, Amaka S, Weinstock DM, and Vardhana SA
- Subjects
- Humans, Middle Aged, Female, Male, Aged, Adult, Aged, 80 and over, Maximum Tolerated Dose, Isoquinolines, Purines, Depsipeptides adverse effects, Depsipeptides therapeutic use, Depsipeptides administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Lymphoma, T-Cell drug therapy, Lymphoma, T-Cell pathology, Bortezomib therapeutic use, Bortezomib administration & dosage, Bortezomib adverse effects
- Abstract
PI3K-δ inhibitors have shown impressive activity in lymphoid malignancies but have been hampered by autoimmune and infectious toxicities, leading to market withdrawals. We previously demonstrated activity of the PI3K-δγ inhibitor duvelisib in T cell lymphomas (TCLs) that was associated with inflammatory adverse events. As reported here, we conducted a phase 1b/2a study of duvelisib in combination with either romidepsin (n = 66) or bortezomib (n = 32) in patients with relapsed/refractory TCL and found that the addition of romidepsin, but not bortezomib, appeared to increase efficacy while attenuating PI3K inhibitor-driven toxicity. The primary endpoint of the study was to determine the safety and maximum tolerated dose of duvelisib, which was 75 mg twice daily when combined with romidepsin versus 25 mg twice daily when combined with bortezomib. The most common adverse events were neutropenia (42%, 25/59) and fatigue (37%, 22/59) in patients treated with duvelisib and romidepsin and diarrhea (48%, 11/23) and neutropenia (30%, 7/23) in patients treated with duvelisib and bortezomib. Duvelisib and romidepsin resulted in less grade 3/4 hepatotoxicity (14%, 8/59) compared to 40% (14/35) in our previous study with duvelisib monotherapy. This was associated with reductions in circulating inflammatory mediators and myeloid cell inflammatory gene expression. Secondary endpoints of overall and complete response rates were 55% (35/64) and 34% (22/64) for patients treated with duvelisib and romidepsin and 34% (11/32) and 13% (4/32) for patients treated with duvelisib and bortezomib. Among patients with peripheral T cell lymphomas (PTCLs), overall and complete response rates of duvelisib and romidepsin were 56% (27/48) and 44% (21/48), respectively, with exploratory analyses showing increased response rates in patients with a follicular helper T cell subtype. These findings support further development of combined PI3K and histone deacetylase (HDAC) inhibition in TCLs and suggest a unique strategy to enable PI3K inhibitor-based combinations for additional patient populations. ClinicalTrials.gov identifier: NCT02783625 ., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
7. Geographic EBV variants confound disease-specific variant interpretation and predict variable immune therapy responses.
- Author
-
Briercheck EL, Ravishankar S, Ahmed EH, Carías Alvarado CC, Barrios Menéndez JC, Silva O, Solórzano-Ortiz E, Siliézar Tala MM, Stevenson P, Xu Y, Wohns AW, Enriquez-Vera D, Barrionuevo C, Yu SC, Freud AG, Oakes C, Weigel C, Weinstock DM, Klimaszewski HL, Ngankeu A, Mutalima N, Samayoa-Reyes G, Newton R, Rochford R, Valvert F, Natkunam Y, Shustov A, Baiocchi RA, and Warren EH
- Subjects
- Humans, Genetic Variation, Genome, Viral, Immunotherapy, Herpesvirus 4, Human genetics, Epstein-Barr Virus Infections immunology
- Abstract
Abstract: Epstein-Barr virus (EBV) is a potent carcinogen linked to hematologic and solid malignancies and causes significant global morbidity and mortality. Therapy using allogeneic EBV-specific lymphocytes shows promise in certain populations, but the impact of EBV genome variation on these strategies remains unexplored. To address this, we sequenced 217 EBV genomes, including hematologic malignancies from Guatemala, Peru, Malawi, and Taiwan, and analyzed them alongside 1307 publicly available EBV genomes from cancer, nonmalignant diseases, and healthy individuals across Africa, Asia, Europe, North America, and South America. These included, to our knowledge, the first natural killer (NK)/T-cell lymphoma (NKTCL) EBV genomes reported outside of East Asia. Our findings indicate that previously proposed EBV genome variants specific to certain cancer types are more closely tied to geographic origin than to cancer histology. This included variants previously reported to be specific to NKTCL but were prevalent in EBV genomes from other cancer types and healthy individuals in East Asia. After controlling for geographic region, we did identify multiple NKTCL-specific variants associated with a 7.8-fold to 21.9-fold increased risk. We also observed frequent variations in EBV genomes that affected peptide sequences previously reported to bind common major histocompatibility complex alleles. Finally, we found several nonsynonymous variants spanning the coding sequences of current vaccine targets BALF4, BKRF2, BLLF1, BXLF2, BZLF1, and BZLF2. These results highlight the need to consider geographic variation in EBV genomes when devising strategies for exploiting adaptive immune responses against EBV-related cancers, ensuring greater global effectiveness and equity in prevention and treatment., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Mutation and cell state compatibility is required and targetable in Ph+ acute lymphoblastic leukemia minimal residual disease.
- Author
-
Winter PS, Ramseier ML, Navia AW, Saksena S, Strouf H, Senhaji N, DenAdel A, Mirza M, An HH, Bilal L, Dennis P, Leahy CS, Shigemori K, Galves-Reyes J, Zhang Y, Powers F, Mulugeta N, Gupta AJ, Calistri N, Van Scoyk A, Jones K, Liu H, Stevenson KE, Ren S, Luskin MR, Couturier CP, Amini AP, Raghavan S, Kimmerling RJ, Stevens MM, Crawford L, Weinstock DM, Manalis SR, Shalek AK, and Murakami MA
- Abstract
Efforts to cure BCR::ABL1 B cell acute lymphoblastic leukemia (Ph+ ALL) solely through inhibition of ABL1 kinase activity have thus far been insufficient despite the availability of tyrosine kinase inhibitors (TKIs) with broad activity against resistance mutants. The mechanisms that drive persistence within minimal residual disease (MRD) remain poorly understood and therefore untargeted. Utilizing 13 patient-derived xenograft (PDX) models and clinical trial specimens of Ph+ ALL, we examined how genetic and transcriptional features co-evolve to drive progression during prolonged TKI response. Our work reveals a landscape of cooperative mutational and transcriptional escape mechanisms that differ from those causing resistance to first generation TKIs. By analyzing MRD during remission, we show that the same resistance mutation can either increase or decrease cellular fitness depending on transcriptional state. We further demonstrate that directly targeting transcriptional state-associated vulnerabilities at MRD can overcome BCR::ABL1 independence, suggesting a new paradigm for rationally eradicating MRD prior to relapse. Finally, we illustrate how cell mass measurements of leukemia cells can be used to rapidly monitor dominant transcriptional features of Ph+ ALL to help rationally guide therapeutic selection from low-input samples., Competing Interests: DECLARATION OF INTERESTS S.R.M., R.J.K., M.M.S., and D.M.W. disclose equity ownership in Travera. A.K.S. reports compensation for consulting and/or SAB membership from Honeycomb Biotechnologies, Cellarity, Bio-Rad Laboratories, Fog Pharma, Passkey Therapeutics, Ochre Bio, Relation Therapeutics, IntrECate biotherapeutics, and Dahlia Biosciences unrelated to this work. P.S.W receives research funding from Microsoft. S.R. holds equity in Amgen and receives research funding from Microsoft. D.M.W. is an employee of Merck and Co., owns equity in Merck and Co., Bantam, Ajax, and Travera, received consulting fees from Astra Zeneca, Secura, Novartis, and Roche/Genentech, and received research support from Daiichi Sankyo, Astra Zeneca, Verastem, Abbvie, Novartis, Abcura, and Surface Oncology. P.S.W., A.K.S., M.A.M., S.R.M., and D.M.W. have filed a patent related to this work. Other authors – none.
- Published
- 2024
- Full Text
- View/download PDF
9. KLRG1 Cell Depletion as a Novel Therapeutic Strategy in Patients with Mature T-Cell Lymphoma Subtypes.
- Author
-
Assatova B, Willim R, Trevisani C, Haskett G, Kariya KM, Chopra K, Park SR, Tolstorukov MY, McCabe SM, Duffy J, Louissaint A Jr, Huuhtanen J, Bhattacharya D, Mustjoki S, Koh MJ, Powers F, Morgan EA, Yang L, Pinckney B, Cotton MJ, Crabbe A, Ziemba JB, Brain I, Heavican-Foral TB, Iqbal J, Nemec R, Rider AB, Ford JG, Koh MJ, Scanlan N, Feith DJ, Loughran TP Jr, Kim WS, Choi J, Roels J, Boehme L, Putteman T, Taghon T, Barnes JA, Johnson PC, Jacobsen ED, Greenberg SA, Weinstock DM, and Jain S
- Subjects
- Animals, Humans, Mice, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal pharmacology, Cell Line, Tumor, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Lymphoma, T-Cell immunology, Lymphoma, T-Cell pathology, Lymphoma, T-Cell therapy, Lymphoma, T-Cell drug therapy, Xenograft Model Antitumor Assays, Lectins, C-Type metabolism, Lectins, C-Type immunology, Lectins, C-Type antagonists & inhibitors, Receptors, Immunologic antagonists & inhibitors, Receptors, Immunologic metabolism, Receptors, Immunologic immunology
- Abstract
Purpose: Develop a novel therapeutic strategy for patients with subtypes of mature T-cell and NK-cell neoplasms., Experimental Design: Primary specimens, cell lines, patient-derived xenograft models, commercially available, and proprietary anti-KLRG1 antibodies were used for screening, target, and functional validation., Results: Here we demonstrate that surface KLRG1 is highly expressed on tumor cells in subsets of patients with extranodal NK/T-cell lymphoma (ENKTCL), T-prolymphocytic leukemia (T-PLL), and gamma/delta T-cell lymphoma (G/D TCL). The majority of the CD8+/CD57+ or CD3-/CD56+ leukemic cells derived from patients with T- and NK-large granular lymphocytic leukemia (T-LGLL and NK-LGLL), respectively, expressed surface KLRG1. The humanized afucosylated anti-KLRG1 monoclonal antibody (mAb208) optimized for mouse in vivo use depleted KLRG1+ TCL cells by mechanisms of ADCC, ADCP, and CDC rather than apoptosis. mAb208 induced ADCC and ADCP of T-LGLL patient-derived CD8+/CD57+ cells ex vivo. mAb208 effected ADCC of subsets of healthy donor-derived KLRG1+ NK, CD4+, CD8+ Tem, and TemRA cells while sparing KLRG1- naïve and CD8+ Tcm cells. Treatment of cell line and TCL patient-derived xenografts with mAb208 or anti-CD47 mAb alone and in combination with the PI3K-δ/γ inhibitor duvelisib extended survival. The depletion of macrophages in vivo antagonized mAb208 efficacy., Conclusions: Our findings suggest the potential benefit of a broader treatment strategy combining therapeutic antibodies with PI3Ki for the treatment of patients with mature T-cell and NK-cell neoplasms. See related commentary by Varma and Diefenbach, p. 2300., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
10. Correction: Chimeric Antigen Receptor T Cells Targeting CD79b Show Efficacy in Lymphoma with or without Cotargeting CD19.
- Author
-
Ormhøj M, Scarfò I, Cabral ML, Bailey SR, Lorrey SJ, Bouffard AA, Castano AP, Larson RC, Riley LS, Schmidts A, Choi BD, Andersen RS, Cédile O, Nyvold CG, Christensen JH, Gjerstorff MF, Ditzel HJ, Weinstock DM, Barington T, Frigault MJ, and Maus MV
- Published
- 2024
- Full Text
- View/download PDF
11. Acquired Multidrug Resistance in AML Is Caused by Low Apoptotic Priming in Relapsed Myeloblasts.
- Author
-
Olesinski EA, Bhatia KS, Wang C, Pioso MS, Lin XX, Mamdouh AM, Ng SX, Sandhu V, Jasdanwala SS, Yilma B, Bohl S, Ryan JA, Malani D, Luskin MR, Kallioniemi O, Porkka K, Adamia S, Chng WJ, Osato M, Weinstock DM, Garcia JS, Letai A, and Bhatt S
- Subjects
- Humans, Animals, Mice, Xenograft Model Antitumor Assays, Granulocyte Precursor Cells drug effects, Granulocyte Precursor Cells pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute genetics, Apoptosis drug effects, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Drug Resistance, Multiple genetics, Drug Resistance, Multiple drug effects
- Abstract
In many cancers, mortality is associated with the emergence of relapse with multidrug resistance (MDR). Thus far, the investigation of cancer relapse mechanisms has largely focused on acquired genetic mutations. Using acute myeloid leukemia (AML) patient-derived xenografts (PDX), we systematically elucidated a basis of MDR and identified drug sensitivity in relapsed AML. We derived pharmacologic sensitivity for 22 AML PDX models using dynamic BH3 profiling (DBP), together with genomics and transcriptomics. Using in vivo acquired resistant PDXs, we found that resistance to unrelated, narrowly targeted agents in distinct PDXs was accompanied by broad resistance to drugs with disparate mechanisms. Moreover, baseline mitochondrial apoptotic priming was consistently reduced regardless of the class of drug-inducing selection. By applying DBP, we identified drugs showing effective in vivo activity in resistant models. This study implies evasion of apoptosis drives drug resistance and demonstrates the feasibility of the DBP approach to identify active drugs for patients with relapsed AML., Significance: Acquired resistance to targeted therapy remains challenging in AML. We found that reduction in mitochondrial priming and common transcriptomic signatures was a conserved mechanism of acquired resistance across different drug classes in vivo. Drugs active in vivo can be identified even in the multidrug resistant state by DBP., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
12. Leukemia circulation kinetics revealed through blood exchange method.
- Author
-
Miller AB, Rodriguez FH, Langenbucher A, Lin L, Bray C, Duquette S, Zhang Y, Goulet D, Lane AA, Weinstock DM, Hemann MT, and Manalis SR
- Subjects
- Mice, Animals, Acute Disease, Vascular Cell Adhesion Molecule-1, Tumor Microenvironment, Bone Marrow pathology, Leukemia, Myeloid, Acute pathology
- Abstract
Leukemias and their bone marrow microenvironments undergo dynamic changes over the course of disease. However, little is known about the circulation kinetics of leukemia cells, nor the impact of specific factors on the clearance of circulating leukemia cells (CLCs) from the blood. To gain a basic understanding of CLC dynamics over the course of disease progression and therapeutic response, we apply a blood exchange method to mouse models of acute leukemia. We find that CLCs circulate in the blood for 1-2 orders of magnitude longer than solid tumor circulating tumor cells. We further observe that: (i) leukemia presence in the marrow can limit the clearance of CLCs in a model of acute lymphocytic leukemia (ALL), and (ii) CLCs in a model of relapsed acute myeloid leukemia (AML) can clear faster than their untreated counterparts. Our approach can also directly quantify the impact of microenvironmental factors on CLC clearance properties. For example, data from two leukemia models suggest that E-selectin, a vascular adhesion molecule, alters CLC clearance. Our research highlights that clearance rates of CLCs can vary in response to tumor and treatment status and provides a strategy for identifying basic processes and factors that govern the kinetics of circulating cells., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Targeting TRIP13 in favorable histology Wilms tumor with nuclear export inhibitors synergizes with doxorubicin.
- Author
-
Mittal K, Cooper GW, Lee BP, Su Y, Skinner KT, Shim J, Jonus HC, Kim WJ, Doshi M, Almanza D, Kynnap BD, Christie AL, Yang X, Cowley GS, Leeper BA, Morton CL, Dwivedi B, Lawrence T, Rupji M, Keskula P, Meyer S, Clinton CM, Bhasin M, Crompton BD, Tseng YY, Boehm JS, Ligon KL, Root DE, Murphy AJ, Weinstock DM, Gokhale PC, Spangle JM, Rivera MN, Mullen EA, Stegmaier K, Goldsmith KC, Hahn WC, and Hong AL
- Subjects
- Humans, Exportin 1 Protein, Active Transport, Cell Nucleus, Karyopherins genetics, Karyopherins metabolism, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear metabolism, Cell Line, Tumor, Apoptosis, Neoplasm Recurrence, Local, Doxorubicin pharmacology, ATPases Associated with Diverse Cellular Activities metabolism, Cell Cycle Proteins metabolism, Wilms Tumor drug therapy, Wilms Tumor genetics, Kidney Neoplasms drug therapy, Kidney Neoplasms genetics, Hydrazines, Triazoles
- Abstract
Wilms tumor (WT) is the most common renal malignancy of childhood. Despite improvements in the overall survival, relapse occurs in ~15% of patients with favorable histology WT (FHWT). Half of these patients will succumb to their disease. Identifying novel targeted therapies remains challenging in part due to the lack of faithful preclinical in vitro models. Here we establish twelve patient-derived WT cell lines and demonstrate that these models faithfully recapitulate WT biology using genomic and transcriptomic techniques. We then perform loss-of-function screens to identify the nuclear export gene, XPO1, as a vulnerability. We find that the FDA approved XPO1 inhibitor, KPT-330, suppresses TRIP13 expression, which is required for survival. We further identify synergy between KPT-330 and doxorubicin, a chemotherapy used in high-risk FHWT. Taken together, we identify XPO1 inhibition with KPT-330 as a potential therapeutic option to treat FHWTs and in combination with doxorubicin, leads to durable remissions in vivo., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. Targeting lysine demethylase 5 (KDM5) in mantle cell lymphoma.
- Author
-
Xu D, Bewicke-Copley F, Close K, Okosun J, Gale RP, Apperley J, Weinstock DM, Wendel HG, and Fitzgibbon J
- Subjects
- Humans, Adult, Histone Demethylases, Lysine, Lymphoma, Mantle-Cell drug therapy
- Published
- 2024
- Full Text
- View/download PDF
15. Targeting Aggressive B-cell Lymphomas through Pharmacological Activation of the Mitochondrial Protease OMA1.
- Author
-
Schwarzer A, Oliveira M, Kleppa MJ, Slattery SD, Anantha A, Cooper A, Hannink M, Schambach A, Dörrie A, Kotlyarov A, Gaestel M, Hembrough T, Levine J, Luther M, Stocum M, Stiles L, Weinstock DM, Liesa M, and Kostura MJ
- Subjects
- Humans, GTP Phosphohydrolases metabolism, Mitochondria metabolism, Mitochondrial Proteins, Peptide Hydrolases metabolism, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell genetics, Lymphoma, B-Cell metabolism
- Abstract
DLBCL are aggressive, rapidly proliferating tumors that critically depend on the ATF4-mediated integrated stress response (ISR) to adapt to stress caused by uncontrolled growth, such as hypoxia, amino acid deprivation, and accumulation of misfolded proteins. Here, we show that ISR hyperactivation is a targetable liability in DLBCL. We describe a novel class of compounds represented by BTM-3528 and BTM-3566, which activate the ISR through the mitochondrial protease OMA1. Treatment of tumor cells with compound leads to OMA1-dependent cleavage of DELE1 and OPA1, mitochondrial fragmentation, activation of the eIF2α-kinase HRI, cell growth arrest, and apoptosis. Activation of OMA1 by BTM-3528 and BTM-3566 is mechanistically distinct from inhibitors of mitochondrial electron transport, as the compounds induce OMA1 activity in the absence of acute changes in respiration. We further identify the mitochondrial protein FAM210B as a negative regulator of BTM-3528 and BTM-3566 activity. Overexpression of FAM210B prevents both OMA1 activation and apoptosis. Notably, FAM210B expression is nearly absent in healthy germinal center B-lymphocytes and in derived B-cell malignancies, revealing a fundamental molecular vulnerability which is targeted by BTM compounds. Both compounds induce rapid apoptosis across diverse DLBCL lines derived from activated B-cell, germinal center B-cell, and MYC-rearranged lymphomas. Once-daily oral dosing of BTM-3566 resulted in complete regression of xenografted human DLBCL SU-DHL-10 cells and complete regression in 6 of 9 DLBCL patient-derived xenografts. BTM-3566 represents a first-of-its kind approach of selectively hyperactivating the mitochondrial ISR for treating DLBCL., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
16. Genetically informed therapy for lymphoma: the discomfiting benefit of lumping splits.
- Author
-
Hahn CK, Palmer AC, and Weinstock DM
- Subjects
- Humans, Rituximab therapeutic use, Cyclophosphamide therapeutic use, Vincristine therapeutic use, Prednisone therapeutic use, Doxorubicin therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Treatment Outcome, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Zhang et al. report a randomized phase 2 trial for diffuse large B cell lymphoma (DLBCL) that compared standard of care (R-CHOP) to R-CHOP combined with one of 5 agents matched to an individual lymphoma's genetics. Overall, the matching strategy significantly outperformed R-CHOP, laying the foundation for a paradigm-shifting phase 3 trial., Competing Interests: Declaration of interests A.C.P. received consulting fees from Merck, Astra Zeneca, and Kymera and research support from Prelude Therapeutics. D.M.W. is an employee of Merck and Co., owns equity in Merck and Co.; Bantam; Ajax; and Travera; received consulting fees from Astra Zeneca; Secura; Novartis; and Roche/Genentech; and received research support from Daiichi Sankyo; Astra Zeneca; Verastem; Abbvie; Novartis; Abcura; and Surface Oncology., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. PD-1 instructs a tumor-suppressive metabolic program that restricts glycolysis and restrains AP-1 activity in T cell lymphoma.
- Author
-
Wartewig T, Daniels J, Schulz M, Hameister E, Joshi A, Park J, Morrish E, Venkatasubramani AV, Cernilogar FM, van Heijster FHA, Hundshammer C, Schneider H, Konstantinidis F, Gabler JV, Klement C, Kurniawan H, Law C, Lee Y, Choi S, Guitart J, Forne I, Giustinani J, Müschen M, Jain S, Weinstock DM, Rad R, Ortonne N, Schilling F, Schotta G, Imhof A, Brenner D, Choi J, and Ruland J
- Subjects
- Mice, Animals, Humans, Transcription Factor AP-1 genetics, Transcription Factor AP-1 metabolism, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Genes, Tumor Suppressor, Acetyl Coenzyme A metabolism, Glycolysis genetics, Lymphoma, T-Cell genetics, Lymphoma, T-Cell, Peripheral
- Abstract
The PDCD1-encoded immune checkpoint receptor PD-1 is a key tumor suppressor in T cells that is recurrently inactivated in T cell non-Hodgkin lymphomas (T-NHLs). The highest frequencies of PDCD1 deletions are detected in advanced disease, predicting inferior prognosis. However, the tumor-suppressive mechanisms of PD-1 signaling remain unknown. Here, using tractable mouse models for T-NHL and primary patient samples, we demonstrate that PD-1 signaling suppresses T cell malignancy by restricting glycolytic energy and acetyl coenzyme A (CoA) production. In addition, PD-1 inactivation enforces ATP citrate lyase (ACLY) activity, which generates extramitochondrial acetyl-CoA for histone acetylation to enable hyperactivity of activating protein 1 (AP-1) transcription factors. Conversely, pharmacological ACLY inhibition impedes aberrant AP-1 signaling in PD-1-deficient T-NHLs and is toxic to these cancers. Our data uncover genotype-specific vulnerabilities in PDCD1-mutated T-NHL and identify PD-1 as regulator of AP-1 activity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. TP63 fusions drive multicomplex enhancer rewiring, lymphomagenesis, and EZH2 dependence.
- Author
-
Wu G, Yoshida N, Liu J, Zhang X, Xiong Y, Heavican-Foral TB, Mandato E, Liu H, Nelson GM, Yang L, Chen R, Donovan KA, Jones MK, Roshal M, Zhang Y, Xu R, Nirmal AJ, Jain S, Leahy C, Jones KL, Stevenson KE, Galasso N, Ganesan N, Chang T, Wu WC, Louissaint A, Debaize L, Yoon H, Dal Cin P, Chan WC, Ho Sui SJ, Ng SY, Feldman AL, Horwitz SM, Adelman K, Fischer ES, Chen CW, Weinstock DM, and Brown M
- Subjects
- Humans, Animals, Mice, Transcriptional Activation, Co-Repressor Proteins, Disease Models, Animal, Enhancer of Zeste Homolog 2 Protein genetics, Transcription Factors, Tumor Suppressor Proteins, Oncogenes, Cell Nucleus
- Abstract
Gene fusions involving tumor protein p63 gene (TP63) occur in multiple T and B cell lymphomas and portend a dismal prognosis for patients. The function and mechanisms of TP63 fusions remain unclear, and there is no target therapy for patients with lymphoma harboring TP63 fusions. Here, we show that TP63 fusions act as bona fide oncogenes and are essential for fusion-positive lymphomas. Transgenic mice expressing TBL1XR1::TP63, the most common TP63 fusion, develop diverse lymphomas that recapitulate multiple human T and B cell lymphomas. Here, we identify that TP63 fusions coordinate the recruitment of two epigenetic modifying complexes, the nuclear receptor corepressor (NCoR)-histone deacetylase 3 (HDAC3) by the N-terminal TP63 fusion partner and the lysine methyltransferase 2D (KMT2D) by the C-terminal TP63 component, which are both required for fusion-dependent survival. TBL1XR1::TP63 localization at enhancers drives a unique cell state that involves up-regulation of MYC and the polycomb repressor complex 2 (PRC2) components EED and EZH2. Inhibiting EZH2 with the therapeutic agent valemetostat is highly effective at treating transgenic lymphoma murine models, xenografts, and patient-derived xenografts harboring TP63 fusions. One patient with TP63 -rearranged lymphoma showed a rapid response to valemetostat treatment. In summary, TP63 fusions link partner components that, together, coordinate multiple epigenetic complexes, resulting in therapeutic vulnerability to EZH2 inhibition.
- Published
- 2023
- Full Text
- View/download PDF
19. Leukemia circulation kinetics revealed through blood exchange method.
- Author
-
Miller AB, Langenbucher A, Rodriguez FH, Lin L, Bray C, Duquette S, Zhang Y, Goulet D, Lane AA, Weinstock DM, Hemann MT, and Manalis SR
- Abstract
Leukemias and their bone marrow microenvironment are known to undergo dynamic changes over the course of disease. However, relatively little is known about the circulation kinetics of leukemia cells, nor the impact of specific factors on the clearance of circulating leukemia cells (CLCs) from the blood. To gain a basic understanding of leukemia cell dynamics over the course of disease progression and therapeutic response, we apply a blood exchange method to mouse models of acute leukemia. We find that CLCs circulate in the blood for 1-2 orders of magnitude longer than solid tumor circulating tumor cells. We further observe that: i) leukemia presence in the marrow can limit the clearance of CLCs in a model of acute lymphocytic leukemia (ALL), and ii) CLCs in a model of relapsed acute myeloid leukemia (AML) can clear faster than their untreated counterparts. Our approach can also directly quantify the impact of microenvironmental factors on CLC clearance properties. For example, data from two leukemia models suggest that E-selectin, a vascular adhesion molecule, alters CLC clearance. Our research highlights that clearance rates of CLCs can vary in response to tumor and treatment status and provides a strategy for identifying basic processes and factors that govern the kinetics of circulating cells.
- Published
- 2023
- Full Text
- View/download PDF
20. Targeting CCR7-PI3Kγ overcomes resistance to tyrosine kinase inhibitors in ALK-rearranged lymphoma.
- Author
-
Mastini C, Campisi M, Patrucco E, Mura G, Ferreira A, Costa C, Ambrogio C, Germena G, Martinengo C, Peola S, Mota I, Vissio E, Molinaro L, Arigoni M, Olivero M, Calogero R, Prokoph N, Tabbò F, Shoji B, Brugieres L, Geoerger B, Turner SD, Cuesta-Mateos C, D'Aliberti D, Mologni L, Piazza R, Gambacorti-Passerini C, Inghirami GG, Chiono V, Kamm RD, Hirsch E, Koch R, Weinstock DM, Aster JC, Voena C, and Chiarle R
- Subjects
- Humans, Animals, Mice, Crizotinib pharmacology, Crizotinib therapeutic use, Receptor Protein-Tyrosine Kinases metabolism, Anaplastic Lymphoma Kinase, Receptors, CCR7 genetics, Tyrosine Kinase Inhibitors, Endothelial Cells metabolism, Phosphatidylinositol 3-Kinases, Protein-Tyrosine Kinases, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Cell Line, Tumor, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Lymphoma, Large-Cell, Anaplastic drug therapy, Lymphoma, Large-Cell, Anaplastic genetics, Lymphoma, Large-Cell, Anaplastic pathology
- Abstract
Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) show potent efficacy in several ALK-driven tumors, but the development of resistance limits their long-term clinical impact. Although resistance mechanisms have been studied extensively in ALK-driven non-small cell lung cancer, they are poorly understood in ALK-driven anaplastic large cell lymphoma (ALCL). Here, we identify a survival pathway supported by the tumor microenvironment that activates phosphatidylinositol 3-kinase γ (PI3K-γ) signaling through the C-C motif chemokine receptor 7 (CCR7). We found increased PI3K signaling in patients and ALCL cell lines resistant to ALK TKIs. PI3Kγ expression was predictive of a lack of response to ALK TKI in patients with ALCL. Expression of CCR7, PI3Kγ, and PI3Kδ were up-regulated during ALK or STAT3 inhibition or degradation and a constitutively active PI3Kγ isoform cooperated with oncogenic ALK to accelerate lymphomagenesis in mice. In a three-dimensional microfluidic chip, endothelial cells that produce the CCR7 ligands CCL19/CCL21 protected ALCL cells from apoptosis induced by crizotinib. The PI3Kγ/δ inhibitor duvelisib potentiated crizotinib activity against ALCL lines and patient-derived xenografts. Furthermore, genetic deletion of CCR7 blocked the central nervous system dissemination and perivascular growth of ALCL in mice treated with crizotinib. Thus, blockade of PI3Kγ or CCR7 signaling together with ALK TKI treatment reduces primary resistance and the survival of persister lymphoma cells in ALCL.
- Published
- 2023
- Full Text
- View/download PDF
21. New scaffolds for type II JAK2 inhibitors overcome the acquired G993A resistance mutation.
- Author
-
Arwood ML, Liu Y, Harkins SK, Weinstock DM, Yang L, Stevenson KE, Plana OD, Dong J, Cirka H, Jones KL, Virtanen AT, Gupta DG, Ceas A, Lawney B, Yoda A, Leahy C, Hao M, He Z, Choi HG, Wang Y, Silvennoinen O, Hubbard SR, Zhang T, Gray NS, and Li LS
- Subjects
- Humans, Mutation, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Myeloproliferative Disorders genetics
- Abstract
Recurrent JAK2 alterations are observed in myeloproliferative neoplasms, B-cell acute lymphoblastic leukemia, and other hematologic malignancies. Currently available type I JAK2 inhibitors have limited activity in these diseases. Preclinical data support the improved efficacy of type II JAK2 inhibitors, which lock the kinase in the inactive conformation. By screening small molecule libraries, we identified a lead compound with JAK2 selectivity. We highlight analogs with on-target biochemical and cellular activity and demonstrate in vivo activity using a mouse model of polycythemia vera. We present a co-crystal structure that confirms the type II binding mode of our compounds with the "DFG-out" conformation of the JAK2 activation loop. Finally, we identify a JAK2 G993A mutation that confers resistance to the type II JAK2 inhibitor CHZ868 but not to our analogs. These data provide a template for identifying novel type II kinase inhibitors and inform further development of agents targeting JAK2 that overcome resistance., Competing Interests: Declaration of interests S.R.H. and O.S. are co-founders, equity holders, and scientific advisory board members of Ajax Therapeutics, Inc. N.S.G. is a founder, science advisory board member (SAB) and equity holder in Syros, C4, Allorion, Lighthorse, Voronoi, Inception, Matchpoint, CobroVentures, GSK, Larkspur (board member), and Soltego (board member). T.Z. is a founder, equity holder, and consultant for Matchpoint. The Gray lab receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Jansen, Kinogen, Arbella, Deerfield, Springworks, Interline, and Sanofi. D.M.W. is an employee of Merck and co-founder and equity holder of Ajax Therapeutics, Inc. and Travera. D.M.W. is an advisor or consultant for Bantam, Ajax Therapeutics, Inc., Secura, Travera, AstraZeneca, and Daiichi Sankyo. The Weinstock lab has received research support from Daiichi Sankyo, Verastem, Secura, and Abcuro. T.Z., Y.L., Z.H., M.H., N.S.G., D.M.W., and L.S.L. are co-inventors on patent number WO 2020/097400 A1 related to this work. K.E.S. is now an employee of Novartis and K.L.J. is now an employee of AstraZeneca., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Integrated multi-omics analyses reveal homology-directed repair pathway as a unique dependency in near-haploid leukemia.
- Author
-
Liu-Lupo Y, Ham JD, Jeewajee SKA, Nguyen L, Delorey T, Ramos A, Weinstock DM, Regev A, and Hemann MT
- Subjects
- Humans, Haploidy, Chromosome Aberrations, DNA Repair, Proteins, Multiomics, Leukemia, Lymphoid
- Abstract
Whole chromosome losses resulting in near-haploid karyotypes are found in a rare subgroup of treatment-refractory acute lymphoblastic leukemia. To systematically dissect the unique physiology and uncover susceptibilities that can be exploited in near-haploid leukemia, we leveraged single-cell RNA-Seq and computational inference of cell cycle stages to pinpoint key differences between near-haploid and diploid leukemia cells. Combining cell cycle stage-specific differential expression with gene essentiality scores from a genome-wide CRISPR-Cas9-mediated knockout screen, we identified the homologous recombination pathway component RAD51B as an essential gene in near-haploid leukemia. DNA damage analyses revealed significantly increased sensitivity of RAD51-mediated repair to RAD51B loss in the G2/M stage of near-haploid cells, suggesting a unique role of RAD51B in the homologous recombination pathway. Elevated G2/M and G1/S checkpoint signaling was part of a RAD51B signature expression program in response to chemotherapy in a xenograft model of human near-haploid B-ALL, and RAD51B and its associated programs were overexpressed in a large panel of near-haploid B-ALL patients. These data highlight a unique genetic dependency on DNA repair machinery in near-haploid leukemia and demarcate RAD51B as a promising candidate for targeted therapy in this treatment-resistant disease., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. ITK degradation to block T cell receptor signaling and overcome therapeutic resistance in T cell lymphomas.
- Author
-
Jiang B, Weinstock DM, Donovan KA, Sun HW, Wolfe A, Amaka S, Donaldson NL, Wu G, Jiang Y, Wilcox RA, Fischer ES, Gray NS, and Wu W
- Subjects
- Humans, Receptors, Antigen, T-Cell metabolism, Drug Resistance, Neoplasm, T-Lymphocytes, Signal Transduction, Lymphoma, T-Cell drug therapy
- Abstract
Interleukin (IL)-2-inducible T cell kinase (ITK) is essential for T cell receptor (TCR) signaling and plays an integral role in T cell proliferation and differentiation. Unlike the ITK homolog BTK, no inhibitors of ITK are currently US Food and Drug Administration (FDA) approved. In addition, recent studies have identified mutations within BTK that confer resistance to both covalent and non-covalent inhibitors. Here, as an alternative strategy, we report the development of BSJ-05-037, a potent and selective heterobifunctional degrader of ITK. BSJ-05-037 displayed enhanced anti-proliferative effects relative to its parent inhibitor BMS-509744, blocked the activation of NF-kB/GATA-3 signaling, and increased the sensitivity of T cell lymphoma cells to cytotoxic chemotherapy both in vitro and in vivo. In summary, targeted degradation of ITK is a novel approach to modulate TCR signal strength that could have broad application for the investigation and treatment of T cell-mediated diseases., Competing Interests: Declaration of interests N.S.G., D.M.W., B.S.J., and W.W. are inventors on a patent application related to the ITK degrader described in this manuscript. N.S.G. is a founder, scientific advisory board (SAB) member, and equity holder in Syros, C4, Allorion, Lighthorse, Voronoi, Inception, Matchpoint, CobroVentures, GSK, Shenandoah (board member), Larkspur (board member), and Soltego (board member). The Gray lab receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Jansen, Kinogen, Arbella, Deerfield, Springworks, Interline, and Sanofi. D.M.W. is an employee of Merck and a founder, SAB member, and equity holder in Ajax and Travera. The Weinstock lab received research funding from AstraZeneca, Daiichi Sankyo, Secura, and Abcuro. E.S.F. is a founder, member of the SAB, and equity holder of Civetta Therapeutics, Jengu Therapeutics, Proximity Therapeutics, and Neomorph Inc.; SAB member and equity holder in Avilar Therapeutics and Photys Therapeutics; and a consultant to Astellas, Sanofi, Novartis, Deerfield, and EcoR1 capital. The Fischer laboratory receives or has received research funding from Novartis, Deerfield, Ajax, Interline, and Astellas. K.A.D. is a consultant to Kronos Bio and Neomorph Inc., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. Assessment of added activity of an antitumor agent.
- Author
-
Chen C, Sun LZ, Ren Y, Rubin EH, Weinstock DM, and Schmidt EV
- Subjects
- Humans, Immunotherapy, Combined Modality Therapy, Antineoplastic Agents therapeutic use, Neoplasms drug therapy
- Abstract
An unprecedented number of novel oncology drugs are under preclinical and clinical development, and nearly all are developed in combinations. With an over-reliance on biological hypotheses, there is less effort to establish single agent activity before initiating late clinical development. This may be contributing to a decreased success rate going from phase 1 to approval in the immunotherapy era. Growing evidence in clinical trial data shows that the treatment benefit from most approved combination therapies can be explained by the independent drug action model. Using this working model, we develop a simple index to measure the added antitumor activity of a new drug based on mean response duration, an endpoint that naturally combines both response status and duration information for all patients, which is shown to be highly predictive of clinical benefit of FDA-approved anti-PD-(L)1 immunotherapies. This index sheds light on challenges and opportunities in contemporary oncology drug development and provides a practical tool to assist with decision-making in early clinical trials., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. Overcoming IMiD resistance in T-cell lymphomas through potent degradation of ZFP91 and IKZF1.
- Author
-
Wu W, Nelson GM, Koch R, Donovan KA, Nowak RP, Heavican-Foral TB, Nirmal AJ, Liu H, Yang L, Duffy J, Powers F, Stevenson KE, Jones MK, Ng SY, Wu G, Jain S, Xu R, Amaka S, Trevisani C, Donaldson NL, Hagner PR, de Leval L, Gaulard P, Iqbal J, Thakurta A, Fischer ES, Adelman K, and Weinstock DM
- Subjects
- Humans, Lenalidomide pharmacology, Thalidomide analogs & derivatives, Thalidomide pharmacology, Ubiquitination, Drug Resistance, Neoplasm, Ikaros Transcription Factor metabolism, Immunologic Factors pharmacology, Lymphoma, T-Cell drug therapy, Multiple Myeloma drug therapy, Ubiquitin-Protein Ligases metabolism
- Abstract
Immunomodulatory (IMiD) agents like lenalidomide and pomalidomide induce the recruitment of IKZF1 and other targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and degradation. These agents are highly active in B-cell lymphomas and a subset of myeloid diseases but have compromised effects in T-cell lymphomas (TCLs). Here, we show that 2 factors determine resistance to IMiDs among TCLs. First, limited CRBN expression reduces IMiD activity in TCLs but can be overcome by newer-generation degrader CC-92480. Using mass spectrometry, we show that CC-92480 selectively degrades IKZF1 and ZFP91 in TCL cells with greater potency than pomalidomide. As a result, CC-92480 is highly active against multiple TCL subtypes and showed greater efficacy than pomalidomide across 4 in vivo TCL models. Second, we demonstrate that ZFP91 functions as a bona fide transcription factor that coregulates cell survival with IKZF1 in IMiD-resistant TCLs. By activating keynote genes from WNT, NF-kB, and MAP kinase signaling, ZFP91 directly promotes resistance to IKZF1 loss. Moreover, lenalidomide-sensitive TCLs can acquire stable resistance via ZFP91 rewiring, which involves casein kinase 2-mediated c-Jun inactivation. Overall, these findings identify a critical transcription factor network within TCLs and provide clinical proof of concept for the novel therapy using next-generation degraders., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
26. A distinct core regulatory module enforces oncogene expression in KMT2A-rearranged leukemia.
- Author
-
Harada T, Heshmati Y, Kalfon J, Perez MW, Xavier Ferrucio J, Ewers J, Hubbell Engler B, Kossenkov A, Ellegast JM, Yi JS, Bowker A, Zhu Q, Eagle K, Liu T, Kai Y, Dempster JM, Kugener G, Wickramasinghe J, Herbert ZT, Li CH, Vrabič Koren J, Weinstock DM, Paralkar VR, Nabet B, Lin CY, Dharia NV, Stegmaier K, Orkin SH, and Pimkin M
- Subjects
- Gene Rearrangement, Humans, Interferon Regulatory Factors genetics, Interferon Regulatory Factors metabolism, Oncogenes genetics, Leukemia, Myeloid, Acute genetics, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism
- Abstract
Acute myeloid leukemia with KMT2A (MLL) rearrangements is characterized by specific patterns of gene expression and enhancer architecture, implying unique core transcriptional regulatory circuitry. Here, we identified the transcription factors MEF2D and IRF8 as selective transcriptional dependencies of KMT2A-rearranged AML, where MEF2D displays partially redundant functions with its paralog, MEF2C. Rapid transcription factor degradation followed by measurements of genome-wide transcription rates and superresolution microscopy revealed that MEF2D and IRF8 form a distinct core regulatory module with a narrow direct transcriptional program that includes activation of the key oncogenes MYC, HOXA9, and BCL2. Our study illustrates a mechanism of context-specific transcriptional addiction whereby a specific AML subclass depends on a highly specialized core regulatory module to directly enforce expression of common leukemia oncogenes., (© 2022 Harada et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
27. The molecular ontogeny of follicular lymphoma: gene mutations succeeding the BCL2 translocation define common precursor cells.
- Author
-
Haebe S, Keay W, Alig S, Mohr AW, Martin LK, Heide M, Secci R, Krebs S, Blum H, Moosmann A, Louissaint A Jr, Weinstock DM, Thoene S, von Bergwelt-Baildon M, Ruland J, Bararia D, and Weigert O
- Subjects
- Hematopoietic Stem Cells metabolism, Humans, Immunoglobulin Heavy Chains genetics, Mutation, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Translocation, Genetic, Lymphoma, B-Cell genetics, Lymphoma, Follicular pathology
- Abstract
Relapsed follicular lymphoma (FL) can arise from common progenitor cells (CPCs). Conceptually, CPC-defining mutations are somatic alterations shared by the initial and relapsed tumours, mostly B-cell leukaemia/lymphoma 2 (BCL2)/immunoglobulin heavy locus (IGH) translocations and other recurrent gene mutations. Through complementary approaches for highly sensitive mutation detection, we do not find CPC-defining mutations in highly purified BCL2/IGH-negative haematopoietic progenitor cells in clinical remission samples from three patients with relapsed FL. Instead, we find cells harbouring the same BCL2/IGH translocation but lacking CREB binding protein (CREBBP), lysine methyltransferase 2D (KMT2D) and other recurrent gene mutations. Thus, (i) the BCL2/IGH translocation can precede CPC-defining mutations in human FL, and (ii) BCL2/IGH-translocated cells can persist in clinical remission., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
28. Identification and Targeting of the Developmental Blockade in Extranodal Natural Killer/T-cell Lymphoma.
- Author
-
Mundy-Bosse BL, Weigel C, Wu YZ, Abdelbaky S, Youssef Y, Casas SB, Polley N, Ernst G, Young KA, McConnell KK, Nalin AP, Wu KG, Broughton M, Lordo MR, Altynova E, Hegewisch-Solloa E, Enriquez-Vera DY, Dueñas D, Barrionuevo C, Yu SC, Saleem A, Suarez CJ, Briercheck EL, Molina-Kirsch H, Loughran TP, Weichenhan D, Plass C, Reneau JC, Mace EM, Gamboa FV, Weinstock DM, Natkunam Y, Caligiuri MA, Mishra A, Porcu P, Baiocchi RA, Brammer JE, Freud AG, and Oakes CC
- Subjects
- Epigenomics, Gene Expression Profiling, Humans, Killer Cells, Natural pathology, Lymphoma, Extranodal NK-T-Cell drug therapy, Natural Killer T-Cells pathology
- Abstract
Extranodal natural killer/T-cell lymphoma (ENKTL) is an aggressive, rare lymphoma of natural killer (NK) cell origin with poor clinical outcomes. Here we used phenotypic and molecular profiling, including epigenetic analyses, to investigate how ENKTL ontogeny relates to normal NK-cell development. We demonstrate that neoplastic NK cells are stably, but reversibly, arrested at earlier stages of NK-cell maturation. Genes downregulated in the most epigenetic immature tumors were associated with polycomb silencing along with genomic gain and overexpression of EZH2. ENKTL cells exhibited genome-wide DNA hypermethylation. Tumor-specific DNA methylation gains were associated with polycomb-marked regions, involving extensive gene silencing and loss of transcription factor binding. To investigate therapeutic targeting, we treated novel patient-derived xenograft (PDX) models of ENKTL with the DNA hypomethylating agent, 5-azacytidine. Treatment led to reexpression of NK-cell developmental genes, phenotypic NK-cell differentiation, and prolongation of survival. These studies lay the foundation for epigenetic-directed therapy in ENKTL., Significance: Through epigenetic and transcriptomic analyses of ENKTL, a rare, aggressive malignancy, along with normal NK-cell developmental intermediates, we identified that extreme DNA hypermethylation targets genes required for NK-cell development. Disrupting this epigenetic blockade in novel PDX models led to ENKTL differentiation and improved survival. This article is highlighted in the In This Issue feature, p. 85., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
29. Sex-Biased ZRSR2 Mutations in Myeloid Malignancies Impair Plasmacytoid Dendritic Cell Activation and Apoptosis.
- Author
-
Togami K, Chung SS, Madan V, Booth CAG, Kenyon CM, Cabal-Hierro L, Taylor J, Kim SS, Griffin GK, Ghandi M, Li J, Li YY, Angelot-Delettre F, Biichle S, Seiler M, Buonamici S, Lovitch SB, Louissaint A Jr, Morgan EA, Jardin F, Piccaluga PP, Weinstock DM, Hammerman PS, Yang H, Konopleva M, Pemmaraju N, Garnache-Ottou F, Abdel-Wahab O, Koeffler HP, and Lane AA
- Subjects
- Apoptosis, Female, Gender Identity, Humans, Male, Mutation, Dendritic Cells metabolism, Myeloproliferative Disorders genetics, Ribonucleoproteins genetics
- Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive leukemia of plasmacytoid dendritic cells (pDC). BPDCN occurs at least three times more frequently in men than in women, but the reasons for this sex bias are unknown. Here, studying genomics of primary BPDCN and modeling disease-associated mutations, we link acquired alterations in RNA splicing to abnormal pDC development and inflammatory response through Toll-like receptors. Loss-of-function mutations in ZRSR2 , an X chromosome gene encoding a splicing factor, are enriched in BPDCN, and nearly all mutations occur in males. ZRSR2 mutation impairs pDC activation and apoptosis after inflammatory stimuli, associated with intron retention and inability to upregulate the transcription factor IRF7. In vivo , BPDCN-associated mutations promote pDC expansion and signatures of decreased activation. These data support a model in which male-biased mutations in hematopoietic progenitors alter pDC function and confer protection from apoptosis, which may impair immunity and predispose to leukemic transformation. SIGNIFICANCE: Sex bias in cancer is well recognized, but the underlying mechanisms are incompletely defined. We connect X chromosome mutations in ZRSR2 to an extremely male-predominant leukemia. Aberrant RNA splicing induced by ZRSR2 mutation impairs dendritic cell inflammatory signaling, interferon production, and apoptosis, revealing a sex- and lineage-related tumor suppressor pathway. This article is highlighted in the In This Issue feature, p. 275 ., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
30. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas.
- Author
-
Moskowitz AJ, Ghione P, Jacobsen E, Ruan J, Schatz JH, Noor S, Myskowski P, Vardhana S, Ganesan N, Hancock H, Davey T, Perez L, Ryu S, Santarosa A, Dowd J, Obadi O, Pomerantz L, Yi N, Sohail S, Galasso N, Neuman R, Liotta B, Blouin W, Baik J, Geyer MB, Noy A, Straus D, Kumar P, Dogan A, Hollmann T, Drill E, Rademaker J, Schoder H, Inghirami G, Weinstock DM, and Horwitz SM
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Janus Kinases antagonists & inhibitors, Lymphoma, T-Cell, Peripheral metabolism, Male, Middle Aged, Molecular Targeted Therapy, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local metabolism, Treatment Outcome, Young Adult, Janus Kinases metabolism, Lymphoma, T-Cell, Peripheral drug therapy, Nitriles therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Pyrimidines therapeutic use, STAT Transcription Factors metabolism
- Abstract
Signaling through JAK1 and/or JAK2 is common among tumor and nontumor cells within peripheral T-cell lymphoma (PTCL). No oral therapies are approved for PTCL, and better treatments for relapsed/refractory disease are urgently needed. We conducted a phase 2 study of the JAK1/2 inhibitor ruxolitinib for patients with relapsed/refractory PTCL (n = 45) or mycosis fungoides (MF) (n = 7). Patients enrolled onto 1 of 3 biomarker-defined cohorts: (1) activating JAK and/or STAT mutations, (2) ≥30% pSTAT3 expression among tumor cells by immunohistochemistry, or (3) neither or insufficient tissue to assess. Patients received ruxolitinib 20 mg PO twice daily until progression and were assessed for response after cycles 2 and 5 and every 3 cycles thereafter. The primary endpoint was clinical benefit rate (CBR), defined as the combination of complete response, partial response (PR), and stable disease lasting at least 6 months. Only 1 of 7 patients with MF had CBR (ongoing PR > 18 months). CBR among the PTCL cases (n = 45) in cohorts 1, 2, and 3 were 53%, 45%, and 13% (cohorts 1 & 2 vs 3, P = .02), respectively. Eight patients had CBR > 12 months (5 ongoing), including 4 of 5 patients with T-cell large granular lymphocytic leukemia. In an exploratory analysis using multiplex immunofluorescence, expression of phosphorylated S6, a marker of PI3 kinase or mitogen-activated protein kinase activation, in <25% of tumor cells was associated with response to ruxolitinib (P = .05). Our findings indicate that ruxolitinib is active across various PTCL subtypes and support a precision therapy approach to JAK/STAT inhibition in patients with PTCL. This trial was registered at www.clincialtrials.gov as #NCT02974647., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Primary cytotoxic T-cell lymphomas harbor recurrent targetable alterations in the JAK-STAT pathway.
- Author
-
Lee K, Evans MG, Yang L, Ng S, Snowden C, Khodadoust M, Brown RA, Trum NA, Querfeld C, Doan LT, Song J, Zhang H, Gru AA, Wood GS, Wada DA, Shanmugam V, Haun PL, Aster JC, Duncan LM, Guitart J, Weinstock DM, Nardi V, and Choi J
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Humans, Janus Kinases genetics, Lymphoma, T-Cell drug therapy, Lymphoma, T-Cell genetics, Molecular Targeted Therapy, Mutation drug effects, STAT Transcription Factors genetics, Janus Kinases metabolism, Lymphoma, T-Cell metabolism, STAT Transcription Factors metabolism, Signal Transduction drug effects
- Published
- 2021
- Full Text
- View/download PDF
32. Low-cost transcriptional diagnostic to accurately categorize lymphomas in low- and middle-income countries.
- Author
-
Valvert F, Silva O, Solórzano-Ortiz E, Puligandla M, Siliézar Tala MM, Guyon T, Dixon SL, López N, López F, Carías Alvarado CC, Terbrueggen R, Stevenson KE, Natkunam Y, Weinstock DM, and Briercheck EL
- Subjects
- Biopsy, Humans, Developing Countries, Lymphoma, T-Cell, Peripheral
- Abstract
Inadequate diagnostics compromise cancer care across lower- and middle-income countries (LMICs). We hypothesized that an inexpensive gene expression assay using paraffin-embedded biopsy specimens from LMICs could distinguish lymphoma subtypes without pathologist input. We reviewed all biopsy specimens obtained at the Instituto de Cancerología y Hospital Dr. Bernardo Del Valle in Guatemala City between 2006 and 2018 for suspicion of lymphoma. Diagnoses were established based on the World Health Organization classification and then binned into 9 categories: nonmalignant, aggressive B-cell, diffuse large B-cell, follicular, Hodgkin, mantle cell, marginal zone, natural killer/T-cell, or mature T-cell lymphoma. We established a chemical ligation probe-based assay (CLPA) that quantifies expression of 37 genes by capillary electrophoresis with reagent/consumable cost of approximately $10/sample. To assign bins based on gene expression, 13 models were evaluated as candidate base learners, and class probabilities from each model were then used as predictors in an extreme gradient boosting super learner. Cases with call probabilities < 60% were classified as indeterminate. Four (2%) of 194 biopsy specimens in storage <3 years experienced assay failure. Diagnostic samples were divided into 70% (n = 397) training and 30% (n = 163) validation cohorts. Overall accuracy for the validation cohort was 86% (95% confidence interval [CI]: 80%-91%). After excluding 28 (17%) indeterminate calls, accuracy increased to 94% (95% CI: 89%-97%). Concordance was 97% for a set of high-probability calls (n = 37) assayed by CLPA in both the United States and Guatemala. Accuracy for a cohort of relapsed/refractory biopsy specimens (n = 39) was 79% and 88%, respectively, after excluding indeterminate cases. Machine-learning analysis of gene expression accurately classifies paraffin-embedded lymphoma biopsy specimens and could transform diagnosis in LMICs., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
33. Combined epigenetic and metabolic treatments overcome differentiation blockade in acute myeloid leukemia.
- Author
-
Zee BM, Poels KE, Yao CH, Kawabata KC, Wu G, Duy C, Jacobus WD, Senior E, Endress JE, Jambhekar A, Lovitch SB, Ma J, Dhall A, Harris IS, Blanco MA, Sykes DB, Licht JD, Weinstock DM, Melnick A, Haigis MC, Michor F, and Shi Y
- Abstract
A hallmark of acute myeloid leukemia (AML) is the inability of self-renewing malignant cells to mature into a non-dividing terminally differentiated state. This differentiation block has been linked to dysregulation of multiple cellular processes, including transcriptional, chromatin, and metabolic regulation. The transcription factor HOXA9 and the histone demethylase LSD1 are examples of such regulators that promote differentiation blockade in AML. To identify metabolic targets that interact with LSD1 inhibition to promote myeloid maturation, we screened a small molecule library to identify druggable substrates. We found that differentiation caused by LSD1 inhibition is enhanced by combined perturbation of purine nucleotide salvage and de novo lipogenesis pathways, and identified multiple lines of evidence to support the specificity of these pathways and suggest a potential basis of how perturbation of these pathways may interact synergistically to promote myeloid differentiation. In sum, these findings suggest potential drug combination strategies in the treatment of AML., Competing Interests: Y.S. is a co-founder and equity holder of Constellation Pharmaceuticals, Athelas Therapeutics and K36 Therapeutics, a consultant for Active Motif, and an equity holder of Imago Biosciences. I.S.H. is a consultant for ONO Pharma USA. D.B.S. is a co-founder and an equity holder of Clear Creek Bio. Y.S., B.M.Z, J.M., and A.D. are developers of the LSD1 inhibitor combinations described in this report and Boston Children's Hospital has pursued a patent application of this technology. All other authors declare no competing financial interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
34. Author Correction: IFITM3 functions as a PIP3 scaffold to amplify PI3K signalling in B cells.
- Author
-
Lee J, Robinson ME, Ma N, Artadji D, Ahmed MA, Xiao G, Sadras T, Deb G, Winchester J, Cosgun KN, Geng H, Chan LN, Kume K, Miettinen TP, Zhang Y, Nix MA, Klemm L, Chen CW, Chen J, Khairnar V, Wiita AP, Thomas-Tikhonenko A, Farzan M, Jung JU, Weinstock DM, Manalis SR, Diamond MS, Vaidehi N, and Müschen M
- Published
- 2021
- Full Text
- View/download PDF
35. Genomic landscape of cutaneous follicular lymphomas reveals 2 subgroups with clinically predictive molecular features.
- Author
-
Zhou XA, Yang J, Ringbloom KG, Martinez-Escala ME, Stevenson KE, Wenzel AT, Fantini D, Martin HK, Moy AP, Morgan EA, Harkins S, Paxton CN, Hong B, Andersen EF, Guitart J, Weinstock DM, Cerroni L, Choi J, and Louissaint A
- Subjects
- Biomarkers, Tumor, Genomics, Humans, Prognosis, Lymphoma, Follicular diagnosis, Lymphoma, Follicular genetics, Skin Neoplasms diagnosis, Skin Neoplasms genetics
- Abstract
Primary cutaneous follicle center lymphomas (PCFCLs) are indolent B-cell lymphomas that predominantly remain skin restricted and manageable with skin-directed therapy. Conversely, secondary cutaneous involvement by usual systemic follicular lymphoma (secondary cutaneous follicular lymphoma [SCFL]) has a worse prognosis and often necessitates systemic therapy. Unfortunately, no histopathologic or genetic features reliably differentiate PCFCL from SCFL at diagnosis. Imaging may miss low-burden internal disease in some cases of SCFLs, leading to misclassification as PCFCL. Whereas usual systemic FL is well characterized genetically, the genomic landscapes of PCFCL and SCFL are unknown. Herein, we analyzed clinicopathologic and immunophenotypic data from 30 cases of PCFCL and 10 of SCFL and performed whole-exome sequencing on 18 specimens of PCFCL and 6 of SCFL. During a median follow-up of 7 years, 26 (87%) of the PCFCLs remained skin restricted. In the remaining 4 cases, systemic disease developed within 3 years of diagnosis. Although the SCFLs universally expressed BCL2 and had BCL2 rearrangements, 73% of the PCFCLs lacked BCL2 expression, and only 8% of skin-restricted PCFCLs had BCL2 rearrangements. SCFLs showed low proliferation fractions, whereas 75% of PCFCLs had proliferation fractions >30%. Of the SCFLs, 67% had characteristic loss-of-function CREBBP or KMT2D mutations vs none in skin-restricted PCFCL. Both SCFL and skin-restricted PCFCL showed frequent TNFRSF14 loss-of-function mutations and copy number loss at chromosome 1p36. These data together establish PCFCL as a unique entity with biological features distinct from usual systemic FL and SCFL. We propose 3 criteria based on BCL2 rearrangement, chromatin-modifying gene mutations (CREBBP, KMT2D, EZH2, and EP300), and proliferation index to classify cutaneous FL specimens based on the likelihood of concurrent or future systemic spread., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
36. RAS-protein activation but not mutation status is an outcome predictor and unifying therapeutic target for high-risk acute lymphoblastic leukemia.
- Author
-
Koschut D, Ray D, Li Z, Giarin E, Groet J, Alić I, Kham SK, Chng WJ, Ariffin H, Weinstock DM, Yeoh AE, Basso G, and Nižetić D
- Subjects
- Animals, Cytokines physiology, Humans, Janus Kinase 2 genetics, Janus Kinase 2 physiology, Mice, Phosphatidylinositol 3-Kinases physiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Tyrosine Phosphatase, Non-Receptor Type 11 physiology, Receptors, Cytokine genetics, Signal Transduction physiology, TOR Serine-Threonine Kinases physiology, ras Proteins antagonists & inhibitors, ras Proteins genetics, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, ras Proteins physiology
- Abstract
Leukemias are routinely sub-typed for risk/outcome prediction and therapy choice using acquired mutations and chromosomal rearrangements. Down syndrome acute lymphoblastic leukemia (DS-ALL) is characterized by high frequency of CRLF2-rearrangements, JAK2-mutations, or RAS-pathway mutations. Intriguingly, JAK2 and RAS-mutations are mutually exclusive in leukemic sub-clones, causing dichotomy in therapeutic target choices. We prove in a cell model that elevated CRLF2 in combination with constitutionally active JAK2 is sufficient to activate wtRAS. On primary clinical DS-ALL samples, we show that wtRAS-activation is an obligatory consequence of mutated/hyperphosphorylated JAK2. We further prove that CRLF2-ligand TSLP boosts the direct binding of active PTPN11 to wtRAS, providing the molecular mechanism for the wtRAS activation. Pre-inhibition of RAS or PTPN11, but not of PI3K or JAK-signaling, prevented TSLP-induced RAS-GTP boost. Cytotoxicity assays on primary clinical DS-ALL samples demonstrated that, regardless of mutation status, high-risk leukemic cells could only be killed using RAS-inhibitor or PTPN11-inhibitor, but not PI3K/JAK-inhibitors, suggesting a unified treatment target for up to 80% of DS-ALL. Importantly, protein activities-based principal-component-analysis multivariate clusters analyzed for independent outcome prediction using Cox proportional-hazards model showed that protein-activity (but not mutation-status) was independently predictive of outcome, demanding a paradigm-shift in patient-stratification strategy for precision therapy in high-risk ALL.
- Published
- 2021
- Full Text
- View/download PDF
37. Reduced Mitochondrial Apoptotic Priming Drives Resistance to BH3 Mimetics in Acute Myeloid Leukemia.
- Author
-
Bhatt S, Pioso MS, Olesinski EA, Yilma B, Ryan JA, Mashaka T, Leutz B, Adamia S, Zhu H, Kuang Y, Mogili A, Louissaint A Jr, Bohl SR, Kim AS, Mehta AK, Sanghavi S, Wang Y, Morris E, Halilovic E, Paweletz CP, Weinstock DM, Garcia JS, and Letai A
- Subjects
- Animals, Apoptosis, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Mice, Mitochondria drug effects, Peptide Fragments pharmacology, Proto-Oncogene Proteins pharmacology, Signal Transduction, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Drug Resistance, Neoplasm, Leukemia, Myeloid, Acute pathology, Mitochondria metabolism, Pyrimidines pharmacology, Sulfonamides pharmacology, Thiophenes pharmacology
- Abstract
Acquired resistance to BH3 mimetic antagonists of BCL-2 and MCL-1 is an important clinical problem. Using acute myelogenous leukemia (AML) patient-derived xenograft (PDX) models of acquired resistance to BCL-2 (venetoclax) and MCL-1 (S63845) antagonists, we identify common principles of resistance and persistent vulnerabilities to overcome resistance. BH3 mimetic resistance is characterized by decreased mitochondrial apoptotic priming as measured by BH3 profiling, both in PDX models and human clinical samples, due to alterations in BCL-2 family proteins that vary among cases, but not to acquired mutations in leukemia genes. BCL-2 inhibition drives sequestered pro-apoptotic proteins to MCL-1 and vice versa, explaining why in vivo combinations of BCL-2 and MCL-1 antagonists are more effective when concurrent rather than sequential. Finally, drug-induced mitochondrial priming measured by dynamic BH3 profiling (DBP) identifies drugs that are persistently active in BH3 mimetic-resistant myeloblasts, including FLT-3 inhibitors and SMAC mimetics., Competing Interests: Declaration of Interests A.L. has consulted for and has received research support from AbbVie, Novartis, and AstraZeneca. He serves on the scientific advisory board of Flash Therapeutics, Dialectic Therapeutics, and Zentalis. J.S.G. received research funding from AbbVie, Genentech, Lilly, and Pfizer. She served on AbbVie advisory board. D.M.W. received research support from Novartis, Abbvie, AstraZeneca, Aileron, Daiichi-Sankyo, and Verastem. He is a founder and equity holder of Ajax Therapeutics and Travera. He serves on scientific advisory boards of Bantam Therapeutics, EDO Mundipharma, and Ajax Therapeutics., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Disagreement, Unenforceability, and Harm Reduction.
- Author
-
Weinstock DM
- Subjects
- Humans, Law Enforcement, Needle-Exchange Programs, Sex Work, Suicide, Assisted, Harm Reduction, Negotiating, Policy
- Abstract
Talk of harm reduction has expanded horizontally, to apply to an ever-widening range of policy domains, and vertically, becoming part of official legal and political discourse. This expansion calls for philosophical theorization. What is the best way in which to characterize harm reduction? Does it represent a distinctive ethical position? How is it best morally justified, and what are its moral limits? I distinguish two varieties of harm reduction. One of them, technocratic harm reduction, is premised on the fact of non-enforceability of prohibitionist policies. The second, deliberative harm reduction, is premised on the fact of reasonable disagreement, grounded in the fact that reasonable persons disagree about a range of controversial behaviours. I argue that deliberative harm reduction better accounts for some of harm reduction's most attractive features, and provides a plausible way of accounting for harm reductions's justificatory grounds and limits.
- Published
- 2020
- Full Text
- View/download PDF
39. IFITM3 functions as a PIP3 scaffold to amplify PI3K signalling in B cells.
- Author
-
Lee J, Robinson ME, Ma N, Artadji D, Ahmed MA, Xiao G, Sadras T, Deb G, Winchester J, Cosgun KN, Geng H, Chan LN, Kume K, Miettinen TP, Zhang Y, Nix MA, Klemm L, Chen CW, Chen J, Khairnar V, Wiita AP, Thomas-Tikhonenko A, Farzan M, Jung JU, Weinstock DM, Manalis SR, Diamond MS, Vaidehi N, and Müschen M
- Subjects
- Animals, Antigens, CD19 metabolism, B-Lymphocytes enzymology, B-Lymphocytes immunology, B-Lymphocytes pathology, Cell Transformation, Neoplastic, Female, Germinal Center cytology, Germinal Center immunology, Germinal Center pathology, Humans, Integrins metabolism, Membrane Microdomains metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Models, Molecular, Phosphorylation, Receptors, Antigen, B-Cell metabolism, B-Lymphocytes metabolism, Membrane Proteins metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphatidylinositol Phosphates metabolism, RNA-Binding Proteins metabolism, Signal Transduction
- Abstract
Interferon-induced transmembrane protein 3 (IFITM3) has previously been identified as an endosomal protein that blocks viral infection
1-3 . Here we studied clinical cohorts of patients with B cell leukaemia and lymphoma, and identified IFITM3 as a strong predictor of poor outcome. In normal resting B cells, IFITM3 was minimally expressed and mainly localized in endosomes. However, engagement of the B cell receptor (BCR) induced both expression of IFITM3 and phosphorylation of this protein at Tyr20, which resulted in the accumulation of IFITM3 at the cell surface. In B cell leukaemia, oncogenic kinases phosphorylate IFITM3 at Tyr20, which causes constitutive localization of this protein at the plasma membrane. In a mouse model, Ifitm3-/- naive B cells developed in normal numbers; however, the formation of germinal centres and the production of antigen-specific antibodies were compromised. Oncogenes that induce the development of leukaemia and lymphoma did not transform Ifitm3-/- B cells. Conversely, the phosphomimetic IFITM3(Y20E) mutant induced oncogenic PI3K signalling and initiated the transformation of premalignant B cells. Mechanistic experiments revealed that IFITM3 functions as a PIP3 scaffold and central amplifier of PI3K signalling. The amplification of PI3K signals depends on IFITM3 using two lysine residues (Lys83 and Lys104) in its conserved intracellular loop as a scaffold for the accumulation of PIP3. In Ifitm3-/- B cells, lipid rafts were depleted of PIP3, which resulted in the defective expression of over 60 lipid-raft-associated surface receptors, and impaired BCR signalling and cellular adhesion. We conclude that the phosphorylation of IFITM3 that occurs after B cells encounter antigen induces a dynamic switch from antiviral effector functions in endosomes to a PI3K amplification loop at the cell surface. IFITM3-dependent amplification of PI3K signalling, which in part acts downstream of the BCR, is critical for the rapid expansion of B cells with high affinity to antigen. In addition, multiple oncogenes depend on IFITM3 to assemble PIP3-dependent signalling complexes and amplify PI3K signalling for malignant transformation.- Published
- 2020
- Full Text
- View/download PDF
40. Polymerase δ promotes chromosomal rearrangements and imprecise double-strand break repair.
- Author
-
Layer JV, Debaize L, Van Scoyk A, House NC, Brown AJ, Liu Y, Stevenson KE, Hemann M, Roberts SA, Price BD, Weinstock DM, and Day TA
- Subjects
- DNA Breaks, Double-Stranded, DNA Polymerase III genetics, Gene Knockdown Techniques, HEK293 Cells, HeLa Cells, Humans, RNA, Small Interfering metabolism, DNA End-Joining Repair, DNA Polymerase III metabolism, Translocation, Genetic
- Abstract
Recent studies have implicated DNA polymerases θ (Pol θ) and β (Pol β) as mediators of alternative nonhomologous end-joining (Alt-NHEJ) events, including chromosomal translocations. Here we identify subunits of the replicative DNA polymerase δ (Pol δ) as promoters of Alt-NHEJ that results in more extensive intrachromosomal mutations at a single double-strand break (DSB) and more frequent translocations between two DSBs. Depletion of the Pol δ accessory subunit POLD2 destabilizes the complex, resulting in degradation of both POLD1 and POLD3 in human cells. POLD2 depletion markedly reduces the frequency of translocations with sequence modifications but does not affect the frequency of translocations with exact joins. Using separation-of-function mutants, we show that both the DNA synthesis and exonuclease activities of the POLD1 subunit contribute to translocations. As described in yeast and unlike Pol θ, Pol δ also promotes homology-directed repair. Codepletion of POLD2 with 53BP1 nearly eliminates translocations. POLD1 and POLD2 each colocalize with phosphorylated H2AX at ionizing radiation-induced DSBs but not with 53BP1. Codepletion of POLD2 with either ligase 3 (LIG3) or ligase 4 (LIG4) does not further reduce translocation frequency compared to POLD2 depletion alone. Together, these data support a model in which Pol δ promotes Alt-NHEJ in human cells at DSBs, including translocations., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
41. DT2216-a Bcl-xL-specific degrader is highly active against Bcl-xL-dependent T cell lymphomas.
- Author
-
He Y, Koch R, Budamagunta V, Zhang P, Zhang X, Khan S, Thummuri D, Ortiz YT, Zhang X, Lv D, Wiegand JS, Li W, Palmer AC, Zheng G, Weinstock DM, and Zhou D
- Subjects
- Aniline Compounds therapeutic use, Aniline Compounds toxicity, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents toxicity, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Blood Platelets drug effects, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cell Line, Tumor, Drug Design, Drug Screening Assays, Antitumor, Drug Synergism, Female, Graft Survival, Humans, Liver pathology, Lymphoma, T-Cell metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Proteins metabolism, Neoplasm Transplantation, Piperazines, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Random Allocation, Spleen pathology, Sulfonamides therapeutic use, Sulfonamides toxicity, Ubiquitin-Protein Ligases chemistry, Xenograft Model Antitumor Assays, bcl-X Protein metabolism, Antineoplastic Agents therapeutic use, Lymphoma, T-Cell drug therapy, Neoplasm Proteins antagonists & inhibitors, bcl-X Protein antagonists & inhibitors
- Abstract
Background: Patients with advanced T cell lymphomas (TCLs) have limited therapeutic options and poor outcomes in part because their TCLs evade apoptosis through upregulation of anti-apoptotic Bcl-2 proteins. Subsets of TCL cell lines, patient-derived xenografts (PDXs), and primary patient samples depend on Bcl-xL for survival. However, small molecule Bcl-xL inhibitors such as ABT263 have failed during clinical development due to on-target and dose-limiting thrombocytopenia., Methods: We have developed DT2216, a proteolysis targeting chimera (PROTAC) targeting Bcl-xL for degradation via Von Hippel-Lindau (VHL) E3 ligase, and shown that it has better anti-tumor activity but is less toxic to platelets compared to ABT263. Here, we examined the therapeutic potential of DT2216 for TCLs via testing its anti-TCL activity in vitro using MTS assay, immunoblotting, and flow cytometry and anti-TCL activity in vivo using TCL cell xenograft and PDX model in mice., Results: The results showed that DT2216 selectively killed various Bcl-xL-dependent TCL cells including MyLa cells in vitro. In vivo, DT2216 alone was highly effective against MyLa TCL xenografts in mice without causing significant thrombocytopenia or other toxicity. Furthermore, DT2216 combined with ABT199 (a selective Bcl-2 inhibitor) synergistically reduced disease burden and improved survival in a TCL PDX mouse model dependent on both Bcl-2 and Bcl-xL., Conclusions: These findings support the clinical testing of DT2216 in patients with Bcl-xL-dependent TCLs, both as a single agent and in rational combinations.
- Published
- 2020
- Full Text
- View/download PDF
42. Signalling input from divergent pathways subverts B cell transformation.
- Author
-
Chan LN, Murakami MA, Robinson ME, Caeser R, Sadras T, Lee J, Cosgun KN, Kume K, Khairnar V, Xiao G, Ahmed MA, Aghania E, Deb G, Hurtz C, Shojaee S, Hong C, Pölönen P, Nix MA, Chen Z, Chen CW, Chen J, Vogt A, Heinäniemi M, Lohi O, Wiita AP, Izraeli S, Geng H, Weinstock DM, and Müschen M
- Subjects
- Animals, B-Lymphocytes pathology, Cell Line, Tumor, Enzyme Activation, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Mice, Protein Tyrosine Phosphatase, Non-Receptor Type 6 metabolism, Proto-Oncogene Proteins c-bcl-6 metabolism, Proto-Oncogene Proteins c-myc metabolism, STAT5 Transcription Factor metabolism, B-Lymphocytes cytology, B-Lymphocytes metabolism, Cell Transformation, Neoplastic, Leukemia, B-Cell metabolism, Leukemia, B-Cell pathology, Signal Transduction
- Abstract
Malignant transformation of cells typically involves several genetic lesions, whose combined activity gives rise to cancer
1 . Here we analyse 1,148 patient-derived B-cell leukaemia (B-ALL) samples, and find that individual mutations do not promote leukaemogenesis unless they converge on one single oncogenic pathway that is characteristic of the differentiation stage of transformed B cells. Mutations that are not aligned with this central oncogenic driver activate divergent pathways and subvert transformation. Oncogenic lesions in B-ALL frequently mimic signalling through cytokine receptors at the pro-B-cell stage (via activation of the signal-transduction protein STAT5)2-4 or pre-B-cell receptors in more mature cells (via activation of the protein kinase ERK)5-8 . STAT5- and ERK-activating lesions are found frequently, but occur together in only around 3% of cases (P = 2.2 × 10-16 ). Single-cell mutation and phospho-protein analyses reveal the segregation of oncogenic STAT5 and ERK activation to competing clones. STAT5 and ERK engage opposing biochemical and transcriptional programs that are orchestrated by the transcription factors MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway comes at the expense of the principal oncogenic driver and reverses transformation. Conversely, deletion of divergent pathway components accelerates leukaemogenesis. Thus, persistence of divergent signalling pathways represents a powerful barrier to transformation, while convergence on one principal driver defines a central event in leukaemia initiation. Pharmacological reactivation of suppressed divergent circuits synergizes strongly with inhibition of the principal oncogenic driver. Hence, reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to enhance treatment responses.- Published
- 2020
- Full Text
- View/download PDF
43. A Novel JAK1 Mutant Breast Implant-Associated Anaplastic Large Cell Lymphoma Patient-Derived Xenograft Fostering Pre-Clinical Discoveries.
- Author
-
Fiore D, Cappelli LV, Zumbo P, Phillips JM, Liu Z, Cheng S, Yoffe L, Ghione P, Di Maggio F, Dogan A, Khodos I, de Stanchina E, Casano J, Kayembe C, Tam W, Betel D, Foa' R, Cerchietti L, Rabadan R, Horwitz S, Weinstock DM, and Inghirami G
- Abstract
Breast implant-associated lymphoma (BIA-ALCL) has recently been recognized as an independent peripheral T-cell lymphoma (PTCL) entity. In this study, we generated the first BIA-ALCL patient-derived tumor xenograft (PDTX) model (IL89) and a matching continuous cell line (IL89_CL#3488) to discover potential vulnerabilities and druggable targets. We characterized IL89 and IL89_CL#3488, both phenotypically and genotypically, and demonstrated that they closely resemble the matching human primary lymphoma. The tumor content underwent significant enrichment along passages, as confirmed by the increased variant allele frequency (VAF) of mutations. Known aberrations (JAK1 and KMT2C) were identified, together with novel hits, including PDGFB, PDGFRA, and SETBP1. A deep sequencing approach allowed the detection of mutations below the Whole Exome Sequencing (WES) sensitivity threshold, including JAK1G1097D, in the primary sample. RNA sequencing confirmed the expression of a signature of differentially expressed genes in BIA-ALCL. Next, we tested IL89's sensitivity to the JAK inhibitor ruxolitinib and observed a potent anti-tumor effect, both in vitro and in vivo. We also implemented a high-throughput drug screening approach to identify compounds associated with increased responses in the presence of ruxolitinib. In conclusion, these new IL89 BIA-ALCL models closely recapitulate the primary correspondent lymphoma and represent an informative platform for dissecting the molecular features of BIA-ALCL and performing pre-clinical drug discovery studies, fostering the development of new precision medicine approaches.
- Published
- 2020
- Full Text
- View/download PDF
44. Cathepsin S Alterations Induce a Tumor-Promoting Immune Microenvironment in Follicular Lymphoma.
- Author
-
Bararia D, Hildebrand JA, Stolz S, Haebe S, Alig S, Trevisani CP, Osorio-Barrios F, Bartoschek MD, Mentz M, Pastore A, Gaitzsch E, Heide M, Jurinovic V, Rautter K, Gunawardana J, Sabdia MB, Szczepanowski M, Richter J, Klapper W, Louissaint A Jr, Ludwig C, Bultmann S, Leonhardt H, Eustermann S, Hopfner KP, Hiddemann W, von Bergwelt-Baildon M, Steidl C, Kridel R, Tobin JWD, Gandhi MK, Weinstock DM, Schmidt-Supprian M, Sárosi MB, Rudelius M, Passerini V, Mautner J, and Weigert O
- Subjects
- Animals, Antigens, Differentiation, B-Lymphocyte metabolism, Cytokines metabolism, Histocompatibility Antigens Class II metabolism, Humans, Immunosuppression Therapy, Lymphoma, Follicular pathology, Mice, Antigen Presentation immunology, Cathepsins metabolism, Lymphoma, Follicular metabolism, Tumor Microenvironment immunology
- Abstract
Tumor cells orchestrate their microenvironment. Here, we provide biochemical, structural, functional, and clinical evidence that Cathepsin S (CTSS) alterations induce a tumor-promoting immune microenvironment in follicular lymphoma (FL). We found CTSS mutations at Y132 in 6% of FL (19/305). Another 13% (37/286) had CTSS amplification, which was associated with higher CTSS expression. CTSS Y132 mutations lead to accelerated autocatalytic conversion from an enzymatically inactive profrom to active CTSS and increased substrate cleavage, including CD74, which regulates major histocompatibility complex class II (MHC class II)-restricted antigen presentation. Lymphoma cells with hyperactive CTSS more efficiently activated antigen-specific CD4
+ T cells in vitro. Tumors with hyperactive CTSS showed increased CD4+ T cell infiltration and proinflammatory cytokine perturbation in a mouse model and in human FLs. In mice, this CTSS-induced immune microenvironment promoted tumor growth. Clinically, patients with CTSS-hyperactive FL had better treatment outcomes with standard immunochemotherapies, indicating that these immunosuppressive regimens target both the lymphoma cells and the tumor-promoting immune microenvironment., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
45. Genomic landscape of young ATLL patients identifies frequent targetable CD28 fusions.
- Author
-
Yoshida N, Shigemori K, Donaldson N, Trevisani C, Cordero NA, Stevenson KE, Bu X, Arakawa F, Takeuchi M, Ohshima K, Yoda A, Ng SY, and Weinstock DM
- Subjects
- Biomarkers, Tumor metabolism, CD28 Antigens metabolism, CTLA-4 Antigen metabolism, Female, Follow-Up Studies, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Leukemia-Lymphoma, Adult T-Cell metabolism, Leukemia-Lymphoma, Adult T-Cell pathology, Male, Middle Aged, Prognosis, Biomarkers, Tumor genetics, CD28 Antigens genetics, CTLA-4 Antigen genetics, Genome, Human, Leukemia-Lymphoma, Adult T-Cell genetics, Mutation, Oncogene Proteins, Fusion genetics
- Abstract
Adult T-cell leukemia/lymphoma (ATLL) in Japan presents at a median age of 70 years and only 5% of patients are <50 years of age. We conducted RNA and targeted DNA sequencing of 8 ATLLs from Japanese patients <50 years of age and identified 3 (37.5%) with both CTLA4-CD28 and inducible costimulator (ICOS)-CD28 fusions. Mutations of PLCG1, PRKCB, and STAT3, which were frequent in other ATLL-sequencing studies, were not identified. Differential expression analysis identified the negative checkpoint molecule LAG3 as the most downregulated gene among cases with the fusions. Immunohistochemistry demonstrated expression of CD80 and CD86, the ligands for CTLA4 and CD28, on ATLL cells and tumor-associated macrophages, respectively. Expression of CTLA4-CD28 in Ba/F3 cells conferred cytokine-independent growth when cocultured with Raji cells that express CD80 and CD86. Growth was associated with recruitment of the p85 subunit of phosphatidylinositol 3-kinase to CTLA4-CD28 and phosphorylation of AKT and extracellular signal-regulated kinase. A CTLA4-blocking antibody reduced cytokine-independent growth in a dose-dependent manner. Together, these results suggest that young Japanese ATLL cases have a unique biology dependent on cell-nonautonomous interactions that drive CD28 signaling. Assessment for CD28 fusions and treatment with CTLA4 blockade should be considered in younger patients with relapsed/refractory ATLL., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
46. Targeting EZH2 for the treatment of hepatosplenic T-cell lymphoma.
- Author
-
Pikman Y, Conway AS, Robichaud AL, Kitara S, Church AJ, Kennedy AL, Silverman LB, Billett AL, Weinstock DM, Harris MH, and Stegmaier K
- Subjects
- Humans, Receptors, Antigen, T-Cell, gamma-delta, Lymphoma, T-Cell drug therapy
- Published
- 2020
- Full Text
- View/download PDF
47. Breaking Down the Barriers to Define and Treat NK/T Cell Lymphoma.
- Author
-
Mundy-Bosse B and Weinstock DM
- Subjects
- Genomics, Humans, Transcriptome, Lymphoma, Lymphoma, T-Cell, Natural Killer T-Cells
- Abstract
In this issue of Cancer Cell, Xiong and colleagues describe the genomic and transcriptional landscape of natural killer/T cell lymphoma (NKTCL), a rare form of non-Hodgkin's lymphoma associated with EBV infection. They divide NKTCL into molecularly defined subtypes that could inform therapeutic strategies for patients with this deadly disease., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and Induces Apoptosis in Hematologic Cancer Cells.
- Author
-
Cidado J, Boiko S, Proia T, Ferguson D, Criscione SW, San Martin M, Pop-Damkov P, Su N, Roamio Franklin VN, Sekhar Reddy Chilamakuri C, D'Santos CS, Shao W, Saeh JC, Koch R, Weinstock DM, Zinda M, Fawell SE, and Drew L
- Subjects
- Apoptosis drug effects, Cyclin-Dependent Kinase 9, Humans, Myeloid Cell Leukemia Sequence 1 Protein, Proteomics, Antineoplastic Agents, Hematologic Neoplasms
- Abstract
Purpose: Cyclin-dependent kinase 9 (CDK9) is a transcriptional regulator and potential therapeutic target for many cancers. Multiple nonselective CDK9 inhibitors have progressed clinically but were limited by a narrow therapeutic window. This work describes a novel, potent, and highly selective CDK9 inhibitor, AZD4573., Experimental Design: The antitumor activity of AZD4573 was determined across broad cancer cell line panels in vitro as well as cell line- and patient-derived xenograft models in vivo . Multiple approaches, including integrated transcriptomic and proteomic analyses, loss-of-function pathway interrogation, and pharmacologic comparisons, were employed to further understand the major mechanism driving AZD4573 activity and to establish an exposure/effect relationship., Results: AZD4573 is a highly selective and potent CDK9 inhibitor. It demonstrated rapid induction of apoptosis and subsequent cell death broadly across hematologic cancer models in vitro , and MCL-1 depletion in a dose- and time-dependent manner was identified as a major mechanism through which AZD4573 induces cell death in tumor cells. This pharmacodynamic (PD) response was also observed in vivo , which led to regressions in both subcutaneous tumor xenografts and disseminated models at tolerated doses both as monotherapy or in combination with venetoclax. This understanding of the mechanism, exposure, and antitumor activity of AZD4573 facilitated development of a robust pharmacokinetic/PD/efficacy model used to inform the clinical trial design., Conclusions: Selective targeting of CDK9 enables the indirect inhibition of MCL-1, providing a therapeutic option for MCL-1-dependent diseases. Accordingly, AZD4573 is currently being evaluated in a phase I clinical trial for patients with hematologic malignancies (clinicaltrials.gov identifier: NCT03263637). See related commentary by Alcon et al., p. 761 ., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
49. Chimeric Antigen Receptor T Cells Targeting CD79b Show Efficacy in Lymphoma with or without Cotargeting CD19.
- Author
-
Ormhøj M, Scarfò I, Cabral ML, Bailey SR, Lorrey SJ, Bouffard AA, Castano AP, Larson RC, Riley LS, Schmidts A, Choi BD, Andersen RS, Cédile O, Nyvold CG, Christensen JH, Gjerstorff MF, Ditzel HJ, Weinstock DM, Barington T, Frigault MJ, and Maus MV
- Subjects
- Animals, Apoptosis, Cell Proliferation, Humans, Lymphocyte Activation, Lymphoma, Mantle-Cell immunology, Lymphoma, Mantle-Cell metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Prognosis, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antigens, CD19 immunology, CD79 Antigens immunology, Immunotherapy, Adoptive methods, Lymphoma, Mantle-Cell therapy, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
Purpose: T cells engineered to express a chimeric antigen receptor (CAR) against CD19 have recently been FDA approved for the treatment of relapsed or refractory large B-cell lymphoma. Despite the success and curative potential of CD19 CAR T cells, several reports describing disease relapse due to antigen loss are now emerging., Experimental Design: We developed a novel CAR construct directed against CD79b, a critical receptor for successful B-cell development that remains highly expressed in several subtypes of B-cell lymphoma, including mantle cell lymphoma (MCL). We tested CAR T cells directed against CD79b alone or in combination with CD19 targeting in a single construct, against cell line- and patient-derived xenograft models., Results: We demonstrate CAR79b antigen-specific recognition and cytotoxicity against a panel of cell lines and patient-derived xenograft models of MCL. Importantly, we show that downregulation of CD19 does not influence surface expression of CD79b and that anti-CD79b CAR T cells alone or arranged in a dual-targeting format with a CD19 single-chain variable fragment (scFv) are able to recognize and eliminate CD19
+ , CD19- , and mixed CD19+ /CD19- B-cell lymphoma., Conclusions: Our findings demonstrate that CAR T cells targeting CD79b alone or in combination have promise for treating and preventing CD19 antigen escape in B-cell lymphomas., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
50. Chemical, Biological, Radiological, Nuclear, and Explosive (CBRNE) Science and the CBRNE Science Medical Operations Science Support Expert (CMOSSE).
- Author
-
Coleman CN, Bader JL, Koerner JF, Hrdina C, Cliffer KD, Hick JL, James JJ, Mansoura MK, Livinski AA, Nystrom SV, DiCarlo-Cohen A, Marinissen MJ, Wathen L, Appler JM, Buddemeier B, Casagrande R, Estes D, Byrne P, Kennedy EM, Jakubowski AA, Case C, Weinstock DM, Dainiak N, Hanfling D, Garrett AL, Grant NN, Dodgen D, Redlener I, MacKAY TF, Treber M, Homer MJ, Taylor TP, Miller A, Korch G, and Hatchett R
- Subjects
- Disaster Planning organization & administration, Disaster Planning trends, Emergency Medical Services trends, Humans, Biohazard Release prevention & control, Chemical Hazard Release prevention & control, Emergency Medical Services methods, Explosive Agents adverse effects, Radioactive Hazard Release prevention & control
- Abstract
A national need is to prepare for and respond to accidental or intentional disasters categorized as chemical, biological, radiological, nuclear, or explosive (CBRNE). These incidents require specific subject-matter expertise, yet have commonalities. We identify 7 core elements comprising CBRNE science that require integration for effective preparedness planning and public health and medical response and recovery. These core elements are (1) basic and clinical sciences, (2) modeling and systems management, (3) planning, (4) response and incident management, (5) recovery and resilience, (6) lessons learned, and (7) continuous improvement. A key feature is the ability of relevant subject matter experts to integrate information into response operations. We propose the CBRNE medical operations science support expert as a professional who (1) understands that CBRNE incidents require an integrated systems approach, (2) understands the key functions and contributions of CBRNE science practitioners, (3) helps direct strategic and tactical CBRNE planning and responses through first-hand experience, and (4) provides advice to senior decision-makers managing response activities. Recognition of both CBRNE science as a distinct competency and the establishment of the CBRNE medical operations science support expert informs the public of the enormous progress made, broadcasts opportunities for new talent, and enhances the sophistication and analytic expertise of senior managers planning for and responding to CBRNE incidents.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.