135 results on '"Guzman ML"'
Search Results
2. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells
- Author
-
Jordan, CT, Upchurch, D, Szilvassy, SJ, Guzman, ML, Howard, DS, Pettigrew, AL, Meyerrose, T, Rossi, R, Grimes, B, Rizzieri, DA, Luger, SM, and Phillips, GL
- Published
- 2000
- Full Text
- View/download PDF
3. A-type proanthocyanidins selectively target acute myeloid leukemia cells in vitro and in vivo
- Author
-
Bystrom, LM, additional, Zong, H, additional, Martinez, LAL, additional, Neto, C, additional, Roboz, GJ, additional, Rivella, S, additional, and Guzman, ML, additional
- Published
- 2016
- Full Text
- View/download PDF
4. Mechanisms of disease: cancer stem cells.
- Author
-
Jordan CT, Guzman ML, and Noble M
- Published
- 2006
5. Cancer stem cells.
- Author
-
Wion D, Berger F, Jordan CT, Guzman ML, and Noble M
- Published
- 2006
6. The co-receptor Neuropilin-1 enhances proliferation in inv(16) acute myeloid leukemia via VEGF signaling.
- Author
-
Hegde M, Ahmad MH, Mulet Lazaro R, Sugita M, Li R, Hu K, Gebhard C, Guzman ML, Bushweller JH, Zhu LJ, Brehm M, Wolfe SA, Delwel R, and Castilla LH
- Abstract
Oncogenic programs regulate the proliferation and maintenance of cancer stem cells, and can define pharmacologic dependencies. In acute myeloid leukemia (AML) with the chromosome inversion 16 (inv(16)), the fusion oncoprotein CBFβ::MYH11 regulates pathways associated with leukemia stem cell activity. Here we demonstrate that expression of Neuropilin-1 (NRP1) is regulated by the fusion oncoprotein, and promotes AML expansion. Mechanistically, we show that the NRP1 locus has open chromatin in inv(16) AML, and that CBFβ::MYH11 modulates the local function of the transcription factors ERG, GATA2 and RUNX1 to sustain NRP1 levels. We found that ERG activates NRP1 expression, and that CBFβ::MYH11 knockdown represses ERG expression, thereby allowing the repressive activity of GATA2/RUNX1 at three NRP1 enhancers. Functionally, we demonstrate that NRP1 enhances the expansion of leukemic cells in vitro and in mice, and that this activity is dependent on its VEGFR-associated FV/FVIII domain. Finally, we show that treatment with VEGF inhibitor axitinib reduces AML cell growth and delays median leukemia latency in vivo. Our findings reveal that the NRP1/VEGF axis mediates proliferation in inv(16) AML blasts, and suggest that targeting NRP1 function could be promising in combination AML therapy., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
7. Multiomic profiling identifies predictors of survival in African American patients with acute myeloid leukemia.
- Author
-
Stiff A, Fornerod M, Kain BN, Nicolet D, Kelly BJ, Miller KE, Mrózek K, Boateng I, Bollas A, Garfinkle EAR, Momoh O, Fasola FA, Olawumi HO, Mencia-Trinchant N, Kloppers JF, van Marle AC, Hu E, Wijeratne S, Wheeler G, Walker CJ, Buss J, Heyrosa A, Desai H, Laganson A, Hamp E, Abu-Shihab Y, Abaza H, Kronen P, Sen S, Johnstone ME, Quinn K, Wronowski B, Hertlein E, Miles LA, Mims AS, Oakes CC, Blachly JS, Larkin KT, Mundy-Bosse B, Carroll AJ, Powell BL, Kolitz JE, Stone RM, Duarte C, Abbott D, Amaya ML, Jordan CT, Uy GL, Stock W, Archer KJ, Paskett ED, Guzman ML, Levine RL, Menghrajani K, Chakravarty D, Berger MF, Bottomly D, McWeeney SK, Tyner JW, Byrd JC, Salomonis N, Grimes HL, Mardis ER, and Eisfeld AK
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Biomarkers, Tumor genetics, Gene Expression Profiling, GTP Phosphohydrolases genetics, Isocitrate Dehydrogenase genetics, Membrane Proteins genetics, Nuclear Proteins genetics, Prognosis, Transcriptome, White genetics, Black or African American genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Mutation, Nucleophosmin
- Abstract
Genomic profiles and prognostic biomarkers in patients with acute myeloid leukemia (AML) from ancestry-diverse populations are underexplored. We analyzed the exomes and transcriptomes of 100 patients with AML with genomically confirmed African ancestry (Black; Alliance) and compared their somatic mutation frequencies with those of 323 self-reported white patients with AML, 55% of whom had genomically confirmed European ancestry (white; BeatAML). Here we find that 73% of 162 gene mutations recurrent in Black patients, including a hitherto unreported PHIP alteration detected in 7% of patients, were found in one white patient or not detected. Black patients with myelodysplasia-related AML were younger than white patients suggesting intrinsic and/or extrinsic dysplasia-causing stressors. On multivariable analyses of Black patients, NPM1 and NRAS mutations were associated with inferior disease-free and IDH1 and IDH2 mutations with reduced overall survival. Inflammatory profiles, cell type distributions and transcriptional profiles differed between Black and white patients with NPM1 mutations. Incorporation of ancestry-specific risk markers into the 2022 European LeukemiaNet genetic risk stratification changed risk group assignment for one-third of Black patients and improved their outcome prediction., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. In Vitro Purging of Acute Lymphoblastic Leukemia (B-ALL) Cells with the Use of PTL, DMAPT, or PU-H71.
- Author
-
Ortiz-Reyes AE, García-Sánchez S, Serrano M, Núñez-Enriquez JC, Alvarado-Moreno JA, Montesinos JJ, Fajardo-Orduña G, Guzman ML, Villasis-Keever MA, Mancilla-Herrera I, Mayani H, and Chavez-Gonzalez A
- Subjects
- Humans, Child, Male, Female, Child, Preschool, Cell Survival drug effects, Antineoplastic Agents pharmacology, Apoptosis drug effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism
- Abstract
Acute lymphoblastic leukemia (ALL) is a hematopoietic disorder that mainly affects the child population, and it is characterized by the presence of lymphoid progenitor or precursor cells with different genetic alterations. The origin of this disease is controversial, since some authors assumed that leukemic transformation occurs in a lymphoid progenitor, and there is also evidence that suggests the existence of leukemic initiating cells (LIC). PTL, DMAPT, and PU-H71 are agents that have been shown to eliminate bulk and stem cells from myeloid leukemias, but this effect has not been analyzed in lymphoblastic leukemias. In this study, we evaluated the effect of these compounds in different populations from pediatric B-ALL. For this, bone marrow samples from pediatric patients without treatment were obtained and cultured in the presence or absence of PTL, DMAPT, and PU-H71. The viability and apoptosis index were analyzed by flow cytometry in different hematopoietic subpopulations. These observations indicate that PTL and DMAPT are able to reduce B-ALL cells with a minimum effect in normal hematopoietic and non-hematopoietic cells. In contrast, PU-H71 was able to reduce the leukemic population and had a minimal effect in normal cells. These results present evidence that PTL and DMAPT are able to abrogate in vitro different populations of B-ALL and could represent a possibility of treatment, as well as prevent disease progression or relapse.
- Published
- 2024
- Full Text
- View/download PDF
9. Immune Checkpoint Inhibitor Therapy and Associations with Clonal Hematopoiesis.
- Author
-
Singh A, Trinchant NM, Mishra R, Arora K, Mehta S, Kuzmanovic T, Zokaei Nikoo M, Singh I, Przespolewski AC, Swaminathan M, Ernstoff MS, Dy GK, Yan L, Sinha E, Sharma S, Hassane DC, Griffiths EA, Wang E, Guzman ML, and Thota S
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Lung Neoplasms genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, DNA-Binding Proteins genetics, Proto-Oncogene Proteins genetics, Adult, Clonal Hematopoiesis genetics, Immune Checkpoint Inhibitors therapeutic use, DNA Methyltransferase 3A, Mutation, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Dioxygenases, Melanoma genetics, Melanoma drug therapy
- Abstract
Cancer cohorts are now known to be associated with increased rates of clonal hematopoiesis (CH). We sort to characterize the hematopoietic compartment of patients with melanoma and non-small cell lung cancer (NSCLC) given our recent population level analysis reporting evolving rates of secondary leukemias. The advent of immune checkpoint blockade (ICB) has dramatically changed our understanding of cancer biology and has altered the standards of care for patients. However, the impact of ICB on hematopoietic myeloid clonal expansion remains to be determined. We studied if exposure to ICB therapy affects hematopoietic clonal architecture and if their evolution contributed to altered hematopoiesis. Blood samples from patients with melanoma and NSCLC ( n = 142) demonstrated a high prevalence of CH. Serial samples (or post ICB exposure samples; n = 25) were evaluated in melanoma and NSCLC patients. Error-corrected sequencing of a targeted panel of genes recurrently mutated in CH was performed on peripheral blood genomic DNA. In serial sample analysis, we observed that mutations in DNMT3A and TET2 increased in size with longer ICB exposures in the melanoma cohort. We also noted that patients with larger size DNMT3A mutations with further post ICB clone size expansion had longer durations of ICB exposure. All serial samples in this cohort showed a statistically significant change in VAF from baseline. In the serial sample analysis of NSCLC patients, we observed similar epigenetic expansion, although not statistically significant. Our study generates a hypothesis for two important questions: (a) Can DNMT3A or TET2 CH serve as predictors of a response to ICB therapy and serve as a novel biomarker of response to ICB therapy? (b) As ICB-exposed patients continue to live longer, the myeloid clonal expansion may portend an increased risk for subsequent myeloid malignancy development. Until now, the selective pressure of ICB/T-cell activating therapies on hematopoietic stem cells were less known and we report preliminary evidence of clonal expansion in epigenetic modifier genes (also referred to as inflammatory CH genes).
- Published
- 2024
- Full Text
- View/download PDF
10. Glucocorticoids paradoxically promote steroid resistance in B cell acute lymphoblastic leukemia through CXCR4/PLC signaling.
- Author
-
Abdoul-Azize S, Hami R, Riou G, Derambure C, Charbonnier C, Vannier JP, Guzman ML, Schneider P, and Boyer O
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Mice, Inbred NOD, Cell Survival drug effects, Receptors, CXCR4 metabolism, Receptors, CXCR4 genetics, Signal Transduction drug effects, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Dexamethasone pharmacology, Type C Phospholipases metabolism, Glucocorticoids pharmacology
- Abstract
Glucocorticoid (GC) resistance in childhood relapsed B-cell acute lymphoblastic leukemia (B-ALL) represents an important challenge. Despite decades of clinical use, the mechanisms underlying resistance remain poorly understood. Here, we report that in B-ALL, GC paradoxically induce their own resistance by activating a phospholipase C (PLC)-mediated cell survival pathway through the chemokine receptor, CXCR4. We identify PLC as aberrantly activated in GC-resistant B-ALL and its inhibition is able to induce cell death by compromising several transcriptional programs. Mechanistically, dexamethasone (Dex) provokes CXCR4 signaling, resulting in the activation of PLC-dependent Ca
2+ and protein kinase C signaling pathways, which curtail anticancer activity. Treatment with a CXCR4 antagonist or a PLC inhibitor improves survival of Dex-treated NSG mice in vivo. CXCR4/PLC axis inhibition significantly reverses Dex resistance in B-ALL cell lines (in vitro and in vivo) and cells from Dex resistant ALL patients. Our study identifies how activation of the PLC signalosome in B-ALL by Dex limits the upfront efficacy of this chemotherapeutic agent., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
11. Cancer microenvironment and pharmacological interventions.
- Author
-
Williams MTS and Guzman ML
- Subjects
- Humans, Tumor Microenvironment, Neoplasms drug therapy
- Published
- 2024
- Full Text
- View/download PDF
12. Understanding the interaction between leukaemia stem cells and their microenvironment to improve therapeutic approaches.
- Author
-
Martinez LM and Guzman ML
- Subjects
- Humans, Stem Cells, Recurrence, Tumor Microenvironment, Neoplasm Recurrence, Local, Leukemia
- Abstract
Although chemotherapeutic regimens can eliminate blasts in leukaemia patients, such therapies are associated with toxicity and often fail to eliminate all malignant cells resulting in disease relapse. Disease relapse has been attributed to the persistence of leukaemia cells in the bone marrow (BM) with the capacity to recapitulate disease; these cells are often referred to as leukaemia stem cells (LSCs). Although LSCs have distinct characteristics in terms of pathobiology and immunophenotype, they are still regulated by their interactions with the surrounding microenvironment. Thus, understanding the interaction between LSCs and their microenvironment is critical to identify effective therapies. To this end, there are numerous efforts to develop models to study such interactions. In this review, we will focus on the reciprocal interactions between LSCs and their milieu in the BM. Furthermore, we will highlight relevant therapies targeting these interactions and discuss some of the promising in vitro models designed to mimic such relationship. LINKED ARTICLES: This article is part of a themed issue on Cancer Microenvironment and Pharmacological Interventions. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.2/issuetoc., (© 2023 British Pharmacological Society.)
- Published
- 2024
- Full Text
- View/download PDF
13. Scalable Strategies to Increase Efficiency and Augment Public Health Activities During Epidemic Peaks.
- Author
-
Pasquale DK, Welsh W, Olson A, Yacoub M, Moody J, Barajas Gomez BA, Bentley-Edwards KL, McCall J, Solis-Guzman ML, Dunn JP, Woods CW, Petzold EA, Bowie AC, Singh K, and Huang ES
- Subjects
- Adult, Humans, Public Health, COVID-19 Testing, SARS-CoV-2, Contact Tracing methods, COVID-19 epidemiology, COVID-19 prevention & control
- Abstract
Objective: Scalable strategies to reduce the time burden and increase contact tracing efficiency are crucial during early waves and peaks of infectious transmission., Design: We enrolled a cohort of SARS-CoV-2-positive seed cases into a peer recruitment study testing social network methodology and a novel electronic platform to increase contact tracing efficiency., Setting: Index cases were recruited from an academic medical center and requested to recruit their local social contacts for enrollment and SARS-CoV-2 testing., Participants: A total of 509 adult participants enrolled over 19 months (384 seed cases and 125 social peers)., Intervention: Participants completed a survey and were then eligible to recruit their social contacts with unique "coupons" for enrollment. Peer participants were eligible for SARS-CoV-2 and respiratory pathogen screening., Main Outcome Measures: The main outcome measures were the percentage of tests administered through the study that identified new SARS-CoV-2 cases, the feasibility of deploying the platform and the peer recruitment strategy, the perceived acceptability of the platform and the peer recruitment strategy, and the scalability of both during pandemic peaks., Results: After development and deployment, few human resources were needed to maintain the platform and enroll participants, regardless of peaks. Platform acceptability was high. Percent positivity tracked with other testing programs in the area., Conclusions: An electronic platform may be a suitable tool to augment public health contact tracing activities by allowing participants to select an online platform for contact tracing rather than sitting for an interview., Competing Interests: J.McCall is an employee of Duke University and serves as a contractor for the US Food and Drug Administration. J.P.D. is a Scientific Advisor at Veri, Inc. C.W.W. is employed by Duke University, Durham Veterans Affairs Healthcare System, and Biomeme; C.W.W. is founder and holds equity in Predigen, Inc; C.W.W. has grants from National Institutes of Health (ARLG, VTEU, NIMHO, NIGMS), DARPA, CDC, PCORI, USAMRAA, DOD, Abbott, Najit, Pfizer, and Sanofi; C.W.W. is a consultant for Arena, Biofire, FHI Clinical, and Karius; C.W.W. is on the advisory board for FHI Clinical and Regeneron, and he is a member of the Board of Directors for Global Health Innovation Alliance Accelerator; C.W.W. serves on a DSMB for Janssen. E.S.H. is an employee of and has equity in Verily Life Sciences, is founder of and has equity in KēlaHealth, and is founder of and has equity in Clinetic. For the remaining authors, no conflicts of interest were declared., (Copyright © 2023 The Authors. Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
14. Systems-level analyses of protein-protein interaction network dysfunctions via epichaperomics identify cancer-specific mechanisms of stress adaptation.
- Author
-
Rodina A, Xu C, Digwal CS, Joshi S, Patel Y, Santhaseela AR, Bay S, Merugu S, Alam A, Yan P, Yang C, Roychowdhury T, Panchal P, Shrestha L, Kang Y, Sharma S, Almodovar J, Corben A, Alpaugh ML, Modi S, Guzman ML, Fei T, Taldone T, Ginsberg SD, Erdjument-Bromage H, Neubert TA, Manova-Todorova K, Tsou MB, Young JC, Wang T, and Chiosis G
- Subjects
- Humans, Proteome metabolism, Protein Interaction Mapping, Acclimatization, Protein Interaction Maps, Neoplasms genetics
- Abstract
Systems-level assessments of protein-protein interaction (PPI) network dysfunctions are currently out-of-reach because approaches enabling proteome-wide identification, analysis, and modulation of context-specific PPI changes in native (unengineered) cells and tissues are lacking. Herein, we take advantage of chemical binders of maladaptive scaffolding structures termed epichaperomes and develop an epichaperome-based 'omics platform, epichaperomics, to identify PPI alterations in disease. We provide multiple lines of evidence, at both biochemical and functional levels, demonstrating the importance of these probes to identify and study PPI network dysfunctions and provide mechanistically and therapeutically relevant proteome-wide insights. As proof-of-principle, we derive systems-level insight into PPI dysfunctions of cancer cells which enabled the discovery of a context-dependent mechanism by which cancer cells enhance the fitness of mitotic protein networks. Importantly, our systems levels analyses support the use of epichaperome chemical binders as therapeutic strategies aimed at normalizing PPI networks., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Updates on radiotherapy-immunotherapy combinations: Proceedings of 6 th annual ImmunoRad conference.
- Author
-
Gregucci F, Spada S, Barcellos-Hoff MH, Bhardwaj N, Chan Wah Hak C, Fiorentino A, Guha C, Guzman ML, Harrington K, Herrera FG, Honeychurch J, Hong T, Iturri L, Jaffee E, Karam SD, Knott SRV, Koumenis C, Lyden D, Marciscano AE, Melcher A, Mondini M, Mondino A, Morris ZS, Pitroda S, Quezada SA, Santambrogio L, Shiao S, Stagg J, Telarovic I, Timmerman R, Vozenin MC, Weichselbaum R, Welsh J, Wilkins A, Xu C, Zappasodi R, Zou W, Bobard A, Demaria S, Galluzzi L, Deutsch E, and Formenti SC
- Subjects
- Humans, Combined Modality Therapy, Immunotherapy, Neoplasms radiotherapy, Neoplasms drug therapy
- Abstract
Focal radiation therapy (RT) has attracted considerable attention as a combinatorial partner for immunotherapy (IT), largely reflecting a well-defined, predictable safety profile and at least some potential for immunostimulation. However, only a few RT-IT combinations have been tested successfully in patients with cancer, highlighting the urgent need for an improved understanding of the interaction between RT and IT in both preclinical and clinical scenarios. Every year since 2016, ImmunoRad gathers experts working at the interface between RT and IT to provide a forum for education and discussion, with the ultimate goal of fostering progress in the field at both preclinical and clinical levels. Here, we summarize the key concepts and findings presented at the Sixth Annual ImmunoRad conference., Competing Interests: MHBH is or has have been a recipient of research grants paid to UCSF or in-kind resources from Roche-Genentech, Varian Medical Systems, Eli Lilly, Pathway Innovations and has received fees for consulting from EMD-Serono, Varian Medical Systems, Genentech, Pathway Innovation, Scholar Rock. KHhas Honoraria: Arch Oncology (Inst), AstraZeneca (Inst), BMS (Inst), Boehringer Ingelheim (Inst), Codiak Biosciences (Inst), F-Star Therapeutics (Inst), Inzen Therapeutics (Inst), Merck Serono (Inst), MSD (Inst), Oncolys Biopharma (Inst), Pfizer (Inst), Replimune (Inst), VacV Biotherapeutics (Inst); Consulting or Advisory Role: Arch Oncology (Inst), AstraZeneca (Inst), BMS (Inst), Boehringer Ingelheim (Inst), Inzen Therapeutics (Inst), Merck Serono (Inst), MSD (Inst), Oncolys BioPharma (Inst), Replimune (Inst); Speakers’ Bureau: BMS (Inst), Merck Serono (Inst), MSD (Inst); Research Funding: AstraZeneca (Inst), Boehringer Ingelheim (Inst), Merck Sharp & Dohme (Inst), Replimune (Inst). FGH received Grant or Research Support Companies from Accuray inc, Bioprotect, Bristol-Myers Squibb, Roche-ImFlame/ImCore, Nanobiotix, AstraZeneca, Debio Pharmaceuticals, Seagen, Eisai, MSD; Grant or Research Support Foundations from Prostate Cancer Foundation, San Salvatore Foundation; Investigator or Co-Investigator Clinical Trials in Bristol-Myers Squibb; Consultations: Johnson & Johnson; Academic Collaborations: EORTC chairman Gynecology Cancer Group, ESMO Scientific Committee member for drug development, ASTRO Scientific Committee Annual Meeting. TH has Consulting: Synthetic Biologics, Novocure, Boston Scientific, Inivata, Merck, GSK; Scientific Advisory Board: PanTher Therapeutics (Equity), Lustgarten; Research Funding (Clinical Trials): Taiho, AstraZeneca, BMS, GSK, IntraOp, Ipsen. EJ reports other support from Abmeta and Adventris, personal fees from Achilles, Dragonfly, Mestag, The Medical Home Group, and Surgtx, other support from Parker Institute, grants and other support from the Lustgarten Foundation, Genentech, BMS, and Break Through Cancer outside the submitted work. SDK receives clinical funding from AstraZeneca, Genentech, and Ionis; she also receives preclinical research funding from Roche. KS is founder and consultant for Faeth Therapeutics and Transomic Technologies. CK is the co-recipient of a Sponsored Research Agreement from Ion Beam Applications (IBA). AM is funded by the Associazione Italiana per la Ricerca sul Cancro (AIRC IG 2018 Id.21763 and AIRC Programma di ricerca 5 per Mille 2019 Id.22737). MM declare grants from Boehringer Ingelheim, AC Biosciences and MSD outside the submitted work. ZSM has Scientific Advisory Board roles and equity options with Archeus Technologies and Seneca Therapeutics. JS owns stock and is a member of the Scientific Advisory Board of Surface Oncology, and is a member of the Scientific Advisory Board of Domain Therapeutics. RT has research grants to his institution from: Varian Medical Systems, Elekta Oncology, Accuray, Inc; scientific advisory board member for: Reflexion Medical, ImmuneSensor Therapeutics. AW acknowledge funding from AstraZeneca and imCORE. RW has stock and other ownership interests with Boost Therapeutics, Immvira LLC, Reflexion Pharmaceuticals, Coordination Pharmaceuticals Inc., Magi Therapeutics, Oncosenescence, Aqualung Therapeutics Corporation, and Cyntegron; he has served in a consulting or advisory role for Aettis Inc., AstraZeneca, Coordination Pharmaceuticals, Genus, Merck Serono S.A., Nano Proteagen, NKGen Biotech, Shuttle Pharmaceuticals, Highlight Therapeutics, S.L., Aqualung Therapeutics Corporation; he has research grants with Varian and Regeneron. RZ is scientific advisory board member of iTeos Therapeutics, receives research grant support from Bristol Myers Squibb and AstraZeneca, and is inventor on patent applications related to work on GITR, CTLA-4, and PD-1 (patent numbers: US20180244793A1; US10323091B2; WO2018106864A1; WO2019094352A1). SD has received compensation for consultant/advisory services from Lytix Biopharma, Mersana Therapeutics, EMD Serono, Ono Pharmaceutical, and Genentech, and research support from Lytix Biopharma and Boehringer-Ingelheim for unrelated projects. LG is/has been holding research contracts with Lytix Biopharma, Promontory and Onxeo, has received consulting/advisory honoraria from Boehringer Ingelheim, AstraZeneca, OmniSEQ, Onxeo, The Longevity Labs, Inzen, Imvax, Sotio, Promontory, Noxopharm, EduCom, and the Luke Heller TECPR2 Foundation, and holds Promontory stock options. ED reports grants and personal fees from Roche Genentech; grants from Servier; grants from AstraZeneca; grants and personal fees from Merck-Serono; grants from BMS; and grants from MSD outside the submitted work. SCF has Consultant: Bayer, Bristol Myers Squibb, Varian, ViewRay, Accuray, Elekta, Janssen, Regeneron, GlaxoSmithKline, Eisai, Astra Zeneca, MedImmune, Merck US, EMD Serono/Merck, Genentech/ROCHE, Boehringer Ingelheim, Nanobiotix and Grant/Research: support from: Bristol Myers Squibb, Varian, Regeneron, Merck, Celldex. All other authors have no conflict of interest to declare., (© 2023 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2023
- Full Text
- View/download PDF
16. Targeted Strategies for Recruitment and Engagement of Latinx Immigrants in Longitudinal Biobehavioral Research.
- Author
-
Mansfield LN, Nagy GA, Solorzano R, Carreño M, Mercado Emerson M, Solis Guzman ML, and Gonzalez-Guarda RM
- Subjects
- Young Adult, Humans, Patient Selection, Ethnicity, Minority Groups, Hispanic or Latino, Trust, Emigrants and Immigrants
- Abstract
Introduction: Racial/ethnic minority communities are underrepresented in research. Medical mistrust and mistreatment, discrimination, and a lack of diverse research workforce may influence recruitment and engagement. Engaging Latinx immigrants for research presents unique recruitment challenges, especially for biobehavioral research which is not well explored. The purpose of this paper is to examine the effect of targeted strategies for recruiting young adult, Latinx immigrants. Methods: Recruitment occurred from 2018 to 2019 in an ongoing, longitudinal, community-engaged research study examining risk and resilience factors for health outcomes in Latinx immigrants. Strategies included active recruitment (e.g., community-based events and public events) and passive recruitment (e.g., word-of-mouth and radio and flyer advertisements). Logistic regression analysis was conducted to examine the influence of type of recruitment on participant enrollment. Results: The study enrolled 391 participants of 701 interested individuals (55%). Greater odds of enrollment were among participants recruited through radio and flyer advertisements (adjusted odds ratio [AOR]=2.90, 95% CI [1.59, 5.27], p =.001), word-of-mouth (AOR=2.50, 95% CI [1.55, 4.03], p <.000), or community-based organization events (AOR=1.68, 95% CI [1.19, 2.38], p =.003). Conclusions: Passive recruitment strategies through trusted sources increased the odds of enrollment of Latinx immigrants in biobehavioral research. Future recruitment efforts should leverage trusted sources to disseminate recruitment materials addressing barriers to recruiting Latinx participants for research.
- Published
- 2023
- Full Text
- View/download PDF
17. Editorial: Measurable residual disease in hematologic malignancies.
- Author
-
Buccisano F, Palmieri R, Guzman ML, and Galimberti S
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
18. Paired bone marrow and peripheral blood samples demonstrate lack of widespread dissemination of some CH clones.
- Author
-
Osman AEG, Mencia-Trinchant N, Saygin C, Moma L, Kim A, Housman G, Pozsgai M, Sinha E, Chandra P, Hassane DC, Sboner A, Sangani K, DiNardi N, Johnson C, Wallace SS, Jabri B, Luu H, Guzman ML, Desai P, and Godley LA
- Subjects
- Humans, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Clone Cells, Bone Marrow, Clonal Hematopoiesis
- Abstract
Clonal hematopoiesis (CH) represents clonal expansion of mutated hematopoietic stem cells detectable in the peripheral blood or bone marrow through next generation sequencing. The current prevailing model posits that CH mutations detected in the peripheral blood mirror bone marrow mutations with clones widely disseminated across hematopoietic compartments. We sought to test the hypothesis that all clones are disseminated throughout hematopoietic tissues by comparing CH in hip vs peripheral blood specimens collected at the time of hip replacement surgery. Here, we show that patients with osteoarthritis have a high prevalence of CH, which involve genes encoding epigenetic modifiers and DNA damage repair pathway proteins. Importantly, we illustrate that CH, including clones with variant allele frequencies >10%, can be confined to specific bone marrow spaces and may be eliminated through surgical excision. Future work will define whether clones with somatic mutations in particular genes or clonal fractions of certain sizes are either more likely to be localized or are slower to disseminate into the peripheral blood and other bony sites., (© 2023 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
- 2023
- Full Text
- View/download PDF
19. Radiation therapy improves CAR T cell activity in acute lymphoblastic leukemia.
- Author
-
Sugita M, Yamazaki T, Alhomoud M, Martinet J, Latouche JB, Golden E, Boyer O, Van Besien K, Formenti SC, Galluzzi L, and Guzman ML
- Subjects
- Humans, Mice, Animals, T-Lymphocytes, Receptors, Antigen, T-Cell, Immunotherapy, Adoptive, Receptors, Chimeric Antigen, Precursor Cell Lymphoblastic Leukemia-Lymphoma radiotherapy, Hematologic Neoplasms
- Abstract
Autologous T cells engineered to express a chimeric antigen receptor (CAR) specific for CD19 are approved for the treatment of various CD19
+ hematological malignancies. While CAR T cells induce objective responses in a majority of patients, relapse frequently occurs upon loss of CD19 expression by neoplastic cells. Radiation therapy (RT) has been successfully employed to circumvent the loss of CAR targets in preclinical models of pancreatic cancer. At least in part, this reflects the ability of RT to elicit death receptor (DR) expression by malignant cells, enabling at least some degree of CAR-independent tumor killing. In a human model of CD19+ acute lymphoblastic leukemia (ALL), we also observed DR upregulation by RT, both in vitro and in vivo. Moreover, low-dose total body irradiation (LD-TBI) delivered to ALL-bearing mice prior to CAR T cell infusion considerably extended the overall survival benefit afforded by CAR T cells alone. Such an improved therapeutic activity was accompanied by a superior expansion of CAR T cells in vivo. These data encourage the initiation of clinical trials combining LD-TBI with CAR T cells in patients with hematological malignancies., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
20. The eukaryotic translation initiation factor eIF4E reprograms alternative splicing.
- Author
-
Ghram M, Morris G, Culjkovic-Kraljacic B, Mars JC, Gendron P, Skrabanek L, Revuelta MV, Cerchietti L, Guzman ML, and Borden KLB
- Subjects
- Humans, RNA Splicing Factors metabolism, Eukaryotic Initiation Factor-4E metabolism, RNA Splicing, Eukaryotic Initiation Factors genetics, Mutation, Alternative Splicing, Leukemia, Myeloid, Acute genetics
- Abstract
Aberrant splicing is typically attributed to splice-factor (SF) mutation and contributes to malignancies including acute myeloid leukemia (AML). Here, we discovered a mutation-independent means to extensively reprogram alternative splicing (AS). We showed that the dysregulated expression of eukaryotic translation initiation factor eIF4E elevated selective splice-factor production, thereby impacting multiple spliceosome complexes, including factors mutated in AML such as SF3B1 and U2AF1. These changes generated a splicing landscape that predominantly supported altered splice-site selection for ~800 transcripts in cell lines and ~4,600 transcripts in specimens from high-eIF4E AML patients otherwise harboring no known SF mutations. Nuclear RNA immunoprecipitations, export assays, polysome analyses, and mutational studies together revealed that eIF4E primarily increased SF production via its nuclear RNA export activity. By contrast, eIF4E dysregulation did not induce known SF mutations or alter spliceosome number. eIF4E interacted with the spliceosome and some pre-mRNAs, suggesting its direct involvement in specific splicing events. eIF4E induced simultaneous effects on numerous SF proteins, resulting in a much larger range of splicing alterations than in the case of mutation or dysregulation of individual SFs and providing a novel paradigm for splicing control and dysregulation., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
21. Editorial: New immunotherapeutic and pharmacological targets and strategies in haematological malignancies.
- Author
-
Williams MTS, Kim YM, and Guzman ML
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
22. Loss of Hepatic Leucine-Rich Repeat-Containing G-Protein Coupled Receptors 4 and 5 Promotes Nonalcoholic Fatty Liver Disease.
- Author
-
Saponara E, Penno C, Orsini V, Wang ZY, Fischer A, Aebi A, Matadamas-Guzman ML, Brun V, Fischer B, Brousseau M, O'Donnell P, Turner J, Graff Meyer A, Bollepalli L, d'Ario G, Roma G, Carbone W, Annunziato S, Obrecht M, Beckmann N, Saravanan C, Osmont A, Tropberger P, Richards SM, Genoud C, Ley S, Ksiazek I, Nigsch F, Terracciano LM, Schadt HS, Bouwmeester T, Tchorz JS, and Ruffner H
- Subjects
- Animals, Mice, beta Catenin metabolism, Leucine metabolism, Liver metabolism, Cholesterol metabolism, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Mice, Inbred C57BL, Diet, High-Fat adverse effects, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
The roof plate-specific spondin-leucine-rich repeat-containing G-protein coupled receptor 4/5 (LGR4/5)-zinc and ring finger 3 (ZNRF3)/ring finger protein 43 (RNF43) module is a master regulator of hepatic Wnt/β-catenin signaling and metabolic zonation. However, its impact on nonalcoholic fatty liver disease (NAFLD) remains unclear. The current study investigated whether hepatic epithelial cell-specific loss of the Wnt/β-catenin modulator Lgr4/5 promoted NAFLD. The 3- and 6-month-old mice with hepatic epithelial cell-specific deletion of both receptors Lgr4/5 (Lgr4/5dLKO) were compared with control mice fed with normal diet (ND) or high-fat diet (HFD). Six-month-old HFD-fed Lgr4/5dLKO mice developed hepatic steatosis and fibrosis but the control mice did not. Serum cholesterol-high-density lipoprotein and total cholesterol levels in 3- and 6-month-old HFD-fed Lgr4/5dLKO mice were decreased compared with those in control mice. An ex vivo primary hepatocyte culture assay and a comprehensive bile acid (BA) characterization in liver, plasma, bile, and feces demonstrated that ND-fed Lgr4/5dLKO mice had impaired BA secretion, predisposing them to develop cholestatic characteristics. Lipidome and RNA-sequencing analyses demonstrated severe alterations in several lipid species and pathways controlling lipid metabolism in the livers of Lgr4/5dLKO mice. In conclusion, loss of hepatic Wnt/β-catenin activity by Lgr4/5 deletion led to loss of BA secretion, cholestatic features, altered lipid homeostasis, and deregulation of lipoprotein pathways. Both BA and intrinsic lipid alterations contributed to the onset of NAFLD., (Copyright © 2022 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
23. Cancer Stem Cells: Biology and Therapeutic Implications.
- Author
-
Mayani H, Chávez-González A, Vázquez-Santillan K, Contreras J, and Guzman ML
- Subjects
- Humans, Neoplastic Stem Cells, Cell Differentiation, Neoplasms drug therapy, Antineoplastic Agents pharmacology
- Abstract
It is well recognized that most cancers derive and progress from transformation and clonal expansion of a single cell that possesses stem cell properties, i.e., self-renewal and multilineage differentiation capacities. Such cancer stem cells (CSCs) are usually present at very low frequencies and possess properties that make them key players in tumor development. Indeed, besides having the ability to initiate tumor growth, CSCs drive tumor progression and metastatic dissemination, are resistant to most cancer drugs, and are responsible for cancer relapse. All of these features make CSCs attractive targets for the development of more effective oncologic treatments. In the present review article, we have summarized recent advances in the biology of CSCs, including their identification through their immunophenotype, and their physiology, both in vivo and in vitro. We have also analyzed some molecular markers that might become targets for developing new therapies aiming at hampering CSCs regeneration and cancer relapse., (Copyright © 2022 Instituto Mexicano del Seguro Social (IMSS). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Fluorescent Multifunctional Organic Nanoparticles for Drug Delivery and Bioimaging: A Tutorial Review.
- Author
-
Vargas-Nadal G, Köber M, Nsamela A, Terenziani F, Sissa C, Pescina S, Sonvico F, Gazzali AM, Wahab HA, Grisanti L, Olivera ME, Palena MC, Guzman ML, Luciani-Giacobbe LC, Jimenez-Kairuz A, Ventosa N, Ratera I, Belfield KD, and Maoz BM
- Abstract
Fluorescent organic nanoparticles (FONs) are a large family of nanostructures constituted by organic components that emit light in different spectral regions upon excitation, due to the presence of organic fluorophores. FONs are of great interest for numerous biological and medical applications, due to their high tunability in terms of composition, morphology, surface functionalization, and optical properties. Multifunctional FONs combine several functionalities in a single nanostructure (emission of light, carriers for drug-delivery, functionalization with targeting ligands, etc.), opening the possibility of using the same nanoparticle for diagnosis and therapy. The preparation, characterization, and application of these multifunctional FONs require a multidisciplinary approach. In this review, we present FONs following a tutorial approach, with the aim of providing a general overview of the different aspects of the design, preparation, and characterization of FONs. The review encompasses the most common FONs developed to date, the description of the most important features of fluorophores that determine the optical properties of FONs, an overview of the preparation methods and of the optical characterization techniques, and the description of the theoretical approaches that are currently adopted for modeling FONs. The last part of the review is devoted to a non-exhaustive selection of some recent biomedical applications of FONs.
- Published
- 2022
- Full Text
- View/download PDF
25. CD123-directed allogeneic chimeric-antigen receptor T-cell therapy (CAR-T) in blastic plasmacytoid dendritic cell neoplasm (BPDCN): Clinicopathological insights.
- Author
-
Pemmaraju N, Wilson NR, Senapati J, Economides MP, Guzman ML, Neelapu SS, Kazemimood R, Davis RE, Jain N, Khoury JD, Sugita M, Cai T, Smith J, Frattini MG, Garton A, Roboz G, and Konopleva M
- Subjects
- Acute Disease, Cytokines metabolism, Dendritic Cells pathology, Humans, Interleukin-3 Receptor alpha Subunit, RNA metabolism, RNA therapeutic use, Hematologic Neoplasms pathology, Hematopoietic Stem Cell Transplantation methods, Myeloproliferative Disorders pathology, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen therapeutic use, Skin Neoplasms metabolism
- Abstract
Purpose: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a hematologic malignancy associated with overexpression of CD123. Allogeneic chimeric antigen receptor T cells (CAR-T) directed against CD123 in BPDCN have been studied in clinical trials. We performed post-mortem analysis of a patient treated with anti-CD123 CAR-T to elucidate cause of death, development of cytokine release syndrome (CRS), and tissue distribution of UCART123 cells., Methods: A post-mortem multidisciplinary clinicopathologic analysis was performed with digital droplet polymerase chain reaction of isolated blood and tissue ribonucleic acid (RNA) to evaluate tissue distribution of infused CAR-T. Multiparameter flow cytometry for detection of CAR-T was used for whole blood samples. Cytokine levels in plasma were measured using multiplex bead assay. Gene expression profiling on isolated RNA was performed using semi-custom Nanostring immune gene panel and RNA-sequence method. RNA in situ hybridization was performed using CAR-specific probe., Results: The patient developed severe clinical CRS refractory to corticosteroids, tocilizumab, and lymphodepletion. Despite significant reduction in BPDCN lesions, the patient passed away on day 9 of CAR-T. Autopsy results show that following lymphodepletion and UCART123 administration, the patient remained severely lymphopenic with few UCART123 cells detected, predominantly localized to spleen., Conclusions: No definitive cause of death was determined, but we hypothesized that the patient may have succumbed to CAR-T-mediated cardiopulmonary toxicity. UCART123 cells displayed low overall distribution, with predominance in immune organs and tissues. Mechanism of CRS development is still poorly understood in patients receiving CAR-T therapy. Future directions in the field developing CD123-targeted agents in BPDCN are discussed., Competing Interests: Disclosure of Conflict of Interest M Konopleva: Consulting/honorarium: AbbVie, Genentech, F. Hoffman La-Roche, Stemline Therapeutics, Amgen, Forty-Seven, and Kisoji. Research funding/clinical trials support: AbbVie, Genentech, F. Hoffman La-Roche, Eli Lilly, Cellectis, Calithera, Ablynx, Stemline Therapeutics, Agios, Ascentage, and Astra Zeneca. Stock options/Royalties: Reata Pharmaceutical. N Pemmaraju: Consulting/honorarium: Celgene, Stemline, Incyte Corporation, Novartis, MustangBio, Roche Diagnostics, and LFB. Research funding/clinical trials support from Stemline, Novartis, Abbvie, Samus, Cellectis, Plexxikon, Daiichi-Sankyo, Affymetrix, and SagerStrong Foundation. M Guzman: Consulting/honorarium: SeqRx. Research funding: AI Therapeutics, TemedOn, Cellectis. S Neelapu: Consulting fees or honorarium from Kite, a Gilead Company, Merck, Bristol Myers Squibb, Novartis, Celgene, Pfizer, Allogene Therapeutics, Athenex, Incyte, Precision Biosciences, Legend Biotech, Adicet Bio, Calibr, Unum Therapeutics, Bluebird Bio, Sana Biotechnology, Caribou, Astellas Pharma, Sellas Life Sciences; research funding from Kite, a Gilead Company, Bristol Myers Squibb, Merck, Poseida, Cellectis, Celgene, Karus Therapeutics, Unum Therapeutics (Cogent Biosciences), Allogene, Precision BioSciences, Acerta and Adicet Bio; royalty income from Takeda Pharmaceuticals; stock options from Longbow Immunotherapy; and patents related to cell therapy. MG Frattini: Employment and Equity ownership: Cellectis, Inc. A Garton: Employment and Equity ownership: Cellectis, Inc. J. Smith: Employment and Equity ownership: Cellectis, S.A., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
26. Ionic complexation improves wound healing in deep second-degree burns and reduces in-vitro ciprofloxacin cytotoxicity in fibroblasts.
- Author
-
Sanchez MF, Guzman ML, Flores-Martín J, Cruz Del Puerto M, Laino C, Soria EA, Donadio AC, Genti-Raimondi S, and Olivera ME
- Subjects
- Alginates pharmacology, Anti-Bacterial Agents pharmacology, Ciprofloxacin pharmacology, Fibroblasts, Humans, Hydrogels pharmacology, Integrin beta1, Ions, Lidocaine, Polyelectrolytes, Wound Healing, Burns drug therapy, Silver Sulfadiazine
- Abstract
The development of new treatments capable of controlling infections and pain related to burns continues to be a challenge. Antimicrobials are necessary tools, but these can be cytotoxic for regenerating cells. In this study, antibiotic-anesthetic (AA) smart systems obtained by ionic complexation of polyelectrolytes with ciprofloxacin and lidocaine were obtained as films and hydrogels. Ionic complexation with sodium alginate and hyaluronate decreased cytotoxicity of ciprofloxacin above 70% in a primary culture of isolated fibroblasts (p < 0.05). In addition, the relative levels of the proteins involved in cell migration, integrin β1 and p-FAK, increased above 1.5 times (p < 0.05) with no significant differences in cell mobility. Evaluation of the systems in a deep second-degree burn model revealed that reepithelization rate was AA-films = AA-hydrogels > control films > no treated > reference cream (silver sulfadiazine cream). In addition, appendage conservation and complete dermis organization were achieved in AA-films and AA-hydrogels. Encouragingly, both the films and the hydrogels showed a significantly superior performance compared to the reference treatment. This work highlights the great potential of this smart system as an attractive dressing for burns, which surpasses currently available treatments., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Analysis of Plant-Plant Interactions Reveals the Presence of Potent Antileukemic Compounds.
- Author
-
Mery DE, Compadre AJ, Ordóñez PE, Selvik EJ, Morocho V, Contreras J, Malagón O, Jones DE, Breen PJ, Balick MJ, Gaudio FG, Guzman ML, and Compadre CM
- Subjects
- Chromatography, Thin Layer, Plant Extracts chemistry, Plant Extracts pharmacology, Plants, Asteraceae chemistry
- Abstract
A method to identify anticancer compounds in plants was proposed based on the hypothesis that these compounds are primarily present in plants to provide them with an ecological advantage over neighboring plants and other competitors. According to this view, identifying plants that contain compounds that inhibit or interfere with the development of other plant species may facilitate the discovery of novel anticancer agents. The method was developed and tested using Magnolia grandiflora , Gynoxys verrucosa , Picradeniopsis oppositifolia , and Hedyosmum racemosum , which are plant species known to possess compounds with cytotoxic activities. Plant extracts were screened for growth inhibitory activity, and then a thin-layer chromatography bioautography assay was conducted. This located the major antileukemic compounds 1 , 2 , 4 , and 5 in the extracts. Once the active compounds were located, they were extracted and purified, and their structures were determined. The growth inhibitory activity of the purified compounds showed a significant correlation with their antileukemic activity. The proposed approach is rapid, inexpensive, and can easily be implemented in areas of the world with high biodiversity but with less access to advanced facilities and biological assays.
- Published
- 2022
- Full Text
- View/download PDF
28. Targeting CD123 in blastic plasmacytoid dendritic cell neoplasm using allogeneic anti-CD123 CAR T cells.
- Author
-
Cai T, Gouble A, Black KL, Skwarska A, Naqvi AS, Taylor D, Zhao M, Yuan Q, Sugita M, Zhang Q, Galetto R, Filipe S, Cavazos A, Han L, Kuruvilla V, Ma H, Weng C, Liu CG, Liu X, Konoplev S, Gu J, Tang G, Su X, Al-Atrash G, Ciurea S, Neelapu SS, Lane AA, Kantarjian H, Guzman ML, Pemmaraju N, Smith J, Thomas-Tikhonenko A, and Konopleva M
- Subjects
- Acute Disease, Animals, Dendritic Cells metabolism, Humans, Interleukin-3 Receptor alpha Subunit metabolism, Mice, Hematologic Neoplasms drug therapy, Hematopoietic Stem Cell Transplantation methods, Myeloproliferative Disorders metabolism, Skin Neoplasms pathology
- Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic malignancy with poor outcomes with conventional therapy. Nearly 100% of BPDCNs overexpress interleukin 3 receptor subunit alpha (CD123). Given that CD123 is differentially expressed on the surface of BPDCN cells, it has emerged as an attractive therapeutic target. UCART123 is an investigational product consisting of allogeneic T cells expressing an anti-CD123 chimeric antigen receptor (CAR), edited with TALEN
® nucleases. In this study, we examine the antitumor activity of UCART123 in preclinical models of BPDCN. We report that UCART123 have selective antitumor activity against CD123-positive primary BPDCN samples (while sparing normal hematopoietic progenitor cells) in the in vitro cytotoxicity and T cell degranulation assays; supported by the increased secretion of IFNγ by UCART123 cells when cultured in the presence of BPDCN cells. UCART123 eradicate BPDCN and result in long-term disease-free survival in a subset of primary patient-derived BPDCN xenograft mouse models. One potential challenge of CD123 targeting therapies is the loss of CD123 antigen through diverse genetic mechanisms, an event observed in one of three BPDCN PDX studied. In summary, these results provide a preclinical proof-of-principle that allogeneic UCART123 cells have potent anti-BPDCN activity., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
29. Allogeneic TCRαβ deficient CAR T-cells targeting CD123 in acute myeloid leukemia.
- Author
-
Sugita M, Galetto R, Zong H, Ewing-Crystal N, Trujillo-Alonso V, Mencia-Trinchant N, Yip W, Filipe S, Lebuhotel C, Gouble A, Hassane DC, Smith J, Roboz GJ, and Guzman ML
- Subjects
- Animals, Humans, Interleukin-3 Receptor alpha Subunit metabolism, Mice, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta metabolism, T-Lymphocytes, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute therapy
- Abstract
Acute myeloid leukemia (AML) is a disease with high incidence of relapse that is originated and maintained from leukemia stem cells (LSCs). Hematopoietic stem cells can be distinguished from LSCs by an array of cell surface antigens such as CD123, thus a candidate to eliminate LSCs using a variety of approaches, including CAR T cells. Here, we evaluate the potential of allogeneic gene-edited CAR T cells targeting CD123 to eliminate LSCs (UCART123). UCART123 cells are TCRαβneg T cells generated from healthy donors using TALEN® gene-editing technology, decreasing the likelihood of graft vs host disease. As safety feature, cells express RQR8 to allow elimination with Rituximab. UCART123 effectively eliminates AML cells in vitro and in vivo with significant benefits in overall survival of AML-patient derived xenograft mice. Furthermore, UCART123 preferentially target AML over normal cells with modest toxicity to normal hematopoietic stem/progenitor cells. Together these results suggest that UCART123 represents an off-the shelf therapeutic approach for AML., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. Methods to monitor in vivo expansion and efficacy of CAR-T cells in preclinical models.
- Author
-
Alhomoud M, Martinet J, Sugita M, Gomez-Arteaga A, and Guzman ML
- Subjects
- Flow Cytometry methods, Polymerase Chain Reaction methods, Immunotherapy, Adoptive methods, T-Lymphocytes
- Abstract
The landscape of CAR-T detection and monitoring techniques in preclinical models is rapidly evolving. In this chapter, we will discuss the most widely used methods. The chapter begins with elaborating on the rational of establishing and optimizing protocols for CAR-T monitoring and explaining why this is a crucial step in CAR-T early development. This conceptual basis will be followed by detailed protocols: polymerase chain reaction (PCR), flow cytometry and bioluminescence imaging (BLI). These in vivo methods can be implemented in labs with interest in CAR-T pre-clinical research. It will provide important tools in the process of CAR-T development and offer better understanding of their efficacy, cytotoxicity and survival., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. Mutant PPM1D - and TP53 -Driven Hematopoiesis Populates the Hematopoietic Compartment in Response to Peptide Receptor Radionuclide Therapy.
- Author
-
Singh A, Mencia-Trinchant N, Griffiths EA, Altahan A, Swaminathan M, Gupta M, Gravina M, Tajammal R, Faber MG, Yan L, Sinha E, Hassane DC, Hayes DN, Guzman ML, Iyer R, Wang ES, and Thota S
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Mutation, Radioisotopes therapeutic use, Radiotherapy adverse effects, Clonal Hematopoiesis genetics, Hematopoiesis, Hematopoietic System, Neuroendocrine Tumors blood, Neuroendocrine Tumors genetics, Neuroendocrine Tumors radiotherapy, Protein Phosphatase 2C genetics, Radioisotopes adverse effects, Receptors, Peptide, Tumor Suppressor Protein p53 physiology
- Abstract
Purpose: Hematologic toxic effects of peptide receptor radionuclide therapy (PRRT) can be permanent. Patients with underlying clonal hematopoiesis (CH) may be more inclined to develop hematologic toxicity after PRRT. However, this association remains understudied., Materials and Methods: We evaluated pre- and post-PRRT blood samples of patients with neuroendocrine tumors. After initial screening, 13 cases of interest were selected. Serial blood samples were obtained on 4 of 13 patients. Genomic DNA was analyzed using a 100-gene panel. A variant allele frequency cutoff of 1% was used to call CH., Result: Sixty-two percent of patients had CH at baseline. Persistent cytopenias were noted in 64% (7 of 11) of the patients. Serial sample analysis demonstrated that PRRT exposure resulted in clonal expansion of mutant DNA damage response genes ( TP53 , CHEK2 , and PPM1D ) and accompanying cytopenias in 75% (3 of 4) of the patients. One patient who had a normal baseline hemogram and developed persistent cytopenias after PRRT exposure showed expansion of mutant PPM1D (variant allele frequency increased to 20% after exposure from < 1% at baseline). In the other two patients, expansion of mutant TP53 , CHEK2 , and PPM1D clones was also noted along with cytopenia development., Conclusion: The shifts in hematopoietic clonal dynamics in our study were accompanied by emergence and persistence of cytopenias. These cytopenias likely represent premalignant state, as PPM1D -, CHEK2 -, and TP53 -mutant clones by themselves carry a high risk for transformation to therapy-related myeloid neoplasms. Future studies should consider CH screening and longitudinal monitoring as a key risk mitigation strategy for patients with neuroendocrine tumors receiving PRRT., Competing Interests: Elizabeth A. GriffithsHonoraria: NovartisConsulting or Advisory Role: Alexion Pharmaceuticals, Takeda, Taiho Oncology, Novartis, Genentech, Celgene/Bristol Myers Squibb, AbbVie, Otsuka US, CTI BioPharma Corp, PicnicHealthResearch Funding: Genentech (Inst), Celgene (Inst), Apellis Pharmaceuticals (Inst), Astex Pharmaceuticals (Inst), Celldex (Inst), Bristol Myers Squibb/CelgeneOpen Payments Link: https://openpaymentsdata.cms.gov/physician/134906 Mark G. FaberStock and Other Ownership Interests: AstraZeneca/MedImmune/Spirogen Eti SinhaEmployment: Foundation MedicineStock and Other Ownership Interests: Roche/Genentech Duane C. HassaneEmployment: TempusStock and Other Ownership Interests: TempusResearch Funding: Daiichi SankyoPatents, Royalties, Other Intellectual Property: ddPCR assessment of minimal residual disease via NPM1 mutations, risk prediction for acute myeloid leukemia David Neil HayesLeadership: GeneCentricStock and Other Ownership Interests: GeneCentricConsulting or Advisory Role: GeneCentric, Merck, Turnstone BioPatents, Royalties, Other Intellectual Property: I hold several diagnostic patents or pending patents in the area of solid tumor diagnostics Monica L. GuzmanConsulting or Advisory Role: SeqRx, Bridge MedicinesResearch Funding: Cellectis (Inst), Bridge Medicines (Inst), Daiichi Sankyo/UCB Japan (Inst) Renuka IyerConsulting or Advisory Role: Lexicon, Novartis, Eisai, Merck, Bayer, Advanced Accelerator Applications, Exelixis, Sun pharma, QED therapeutics, Ipsen, Sandoz, TerSera, AstraZenecaResearch Funding: Genentech/Roche (Inst), Ipsen (Inst), Lilly (Inst), Merck (Inst), Taiho Pharmaceutical (Inst), Taiho Pharmaceutical (Inst), Cleveland BioLabs (Inst), Novartis (Inst) Eunice S. WangConsulting or Advisory Role: AbbVie, Pfizer, Jazz Pharmaceuticals, Astellas Pharma, Stemline Therapeutics, Kite/Gilead, MacroGenics, PTC Therapeutics, Celgene/Bristol Myers Squibb, GlaxoSmithKline, Novartis, Genentech, TakedaSpeakers' Bureau: Stemline Therapeutics, Pfizer, Dava Oncology Swapna ThotaHonoraria: Blueprint Medicines, Incyte, Adelson Medical Research FoundationSpeakers' Bureau: IncyteNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
32. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party.
- Author
-
Heuser M, Freeman SD, Ossenkoppele GJ, Buccisano F, Hourigan CS, Ngai LL, Tettero JM, Bachas C, Baer C, Béné MC, Bücklein V, Czyz A, Denys B, Dillon R, Feuring-Buske M, Guzman ML, Haferlach T, Han L, Herzig JK, Jorgensen JL, Kern W, Konopleva MY, Lacombe F, Libura M, Majchrzak A, Maurillo L, Ofran Y, Philippe J, Plesa A, Preudhomme C, Ravandi F, Roumier C, Subklewe M, Thol F, van de Loosdrecht AA, van der Reijden BA, Venditti A, Wierzbowska A, Valk PJM, Wood BL, Walter RB, Thiede C, Döhner K, Roboz GJ, and Cloos J
- Subjects
- Europe, Flow Cytometry methods, High-Throughput Nucleotide Sequencing methods, Humans, Leukemia, Myeloid, Acute genetics, Neoplasm, Residual genetics, Prognosis, Leukemia, Myeloid, Acute diagnosis, Neoplasm, Residual diagnosis
- Abstract
Measurable residual disease (MRD) is an important biomarker in acute myeloid leukemia (AML) that is used for prognostic, predictive, monitoring, and efficacy-response assessments. The European LeukemiaNet (ELN) MRD Working Party evaluated standardization and harmonization of MRD in an ongoing manner and has updated the 2018 ELN MRD recommendations based on significant developments in the field. New and revised recommendations were established during in-person and online meetings, and a 2-stage Delphi poll was conducted to optimize consensus. All recommendations are graded by levels of evidence and agreement. Major changes include technical specifications for next-generation sequencing-based MRD testing and integrative assessments of MRD irrespective of technology. Other topics include use of MRD as a prognostic and surrogate end point for drug testing; selection of the technique, material, and appropriate time points for MRD assessment; and clinical implications of MRD assessment. In addition to technical recommendations for flow- and molecular-MRD analysis, we provide MRD thresholds and define MRD response, and detail how MRD results should be reported and combined if several techniques are used. MRD assessment in AML is complex and clinically relevant, and standardized approaches to application, interpretation, technical conduct, and reporting are of critical importance., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
33. Technical Aspects of Flow Cytometry-based Measurable Residual Disease Quantification in Acute Myeloid Leukemia: Experience of the European LeukemiaNet MRD Working Party.
- Author
-
Tettero JM, Freeman S, Buecklein V, Venditti A, Maurillo L, Kern W, Walter RB, Wood BL, Roumier C, Philippé J, Denys B, Jorgensen JL, Bene MC, Lacombe F, Plesa A, Guzman ML, Wierzbowska A, Czyz A, Ngai LL, Schwarzer A, Bachas C, Cloos J, Subklewe M, Fuering-Buske M, and Buccisano F
- Abstract
Measurable residual disease (MRD) quantified by multiparameter flow cytometry (MFC) is a strong and independent prognostic factor in acute myeloid leukemia (AML). However, several technical factors may affect the final read-out of the assay. Experts from the MRD Working Party of the European LeukemiaNet evaluated which aspects are crucial for accurate MFC-MRD measurement. Here, we report on the agreement, obtained via a combination of a cross-sectional questionnaire, live discussions, and a Delphi poll. The recommendations consist of several key issues from bone marrow sampling to final laboratory reporting to ensure quality and reproducibility of results. Furthermore, the experiences were tested by comparing two 8-color MRD panels in multiple laboratories. The results presented here underscore the feasibility and the utility of a harmonized theoretical and practical MFC-MRD assessment and are a next step toward further harmonization., (Copyright © 2021 the Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the European Hematology Association.)
- Published
- 2021
- Full Text
- View/download PDF
34. Patient-Derived Bone Marrow Spheroids Reveal Leukemia-Initiating Cells Supported by Mesenchymal Hypoxic Niches in Pediatric B-ALL.
- Author
-
Balandrán JC, Dávila-Velderrain J, Sandoval-Cabrera A, Zamora-Herrera G, Terán-Cerqueda V, García-Stivalet LA, Limón-Flores JA, Armenta-Castro E, Rodríguez-Martínez A, Leon-Chavez BA, Vallejo-Ruiz V, Hassane DC, Pérez-Tapia SM, Ortiz-Navarrete V, Guzman ML, and Pelayo R
- Subjects
- Animals, Bone Marrow pathology, Female, Heterografts, Humans, Mesenchymal Stem Cells pathology, Mice, Tumor Microenvironment, Neoplastic Stem Cells pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Spheroids, Cellular, Stem Cell Niche, Tumor Cells, Cultured
- Abstract
B-cell acute lymphoblastic leukemia (B-ALL) results from the expansion of malignant lymphoid precursors within the bone marrow (BM), where hematopoietic niches and microenvironmental signals provide leukemia-initiating cells (LICs) the conditions to survive, proliferate, initiate disease, and relapse. Normal and malignant lymphopoiesis are highly dependent on the BM microenvironment, particularly on CXCL12-abundant Reticular (CAR) cells, which provide a niche for maintenance of primitive cells. During B-ALL, leukemic cells hijack BM niches, creating a proinflammatory milieu incompetent to support normal hematopoiesis but favoring leukemic proliferation. Although the lack of a phenotypic stem cell hierarchy is apparent in B-ALL, LICs are a rare and quiescent population potentially responsible for chemoresistance and relapse. Here, we developed novel patient-derived leukemia spheroids (PDLS), an ex vivo avatar model, from mesenchymal stromal cells (MSCs) and primary B-ALL cells, to mimic specialized niche structures and cell-to-cell intercommunication promoting normal and malignant hematopoiesis in pediatric B-ALL. 3D MSC spheroids can recapitulate CAR niche-like hypoxic structures that produce high levels of CXCL10 and CXCL11. We found that PDLS were preferentially enriched with leukemia cells displaying functional properties of LICs, such as quiescence, low reactive oxygen species, drug resistance, high engraftment in immunodeficient mice, and long-term leukemogenesis. Moreover, the combination of PDLS and patient-derived xenografts confirmed a microenvironment-driven hierarchy in their leukemic potential. Importantly, transcriptional profiles of MSC derived from primary patient samples revealed two unique signatures (1), a CXCL12
low inflammatory and leukemia expansion (ILE)-like niche, that likely supports leukemic burden, and (2) a CXCL11hi immune-suppressive and leukemia-initiating cell (SLIC)-like niche, where LICs are likely sustained. Interestingly, the CXCL11+ hypoxic zones were recapitulated within the PDLS that are capable of supporting LIC functions. Taken together, we have implemented a novel PDLS system that enriches and supports leukemia cells with stem cell features driven by CXCL11+ MSCs within hypoxic microenvironments capable of recapitulating key features, such as tumor reemergence after exposure to chemotherapy and tumor initiation. This system represents a unique opportunity for designing ex vivo personalized avatars for B-ALL patients to evaluate their own LIC pathobiology and drug sensitivity in the context of the tumor microenvironment., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Balandrán, Dávila-Velderrain, Sandoval-Cabrera, Zamora-Herrera, Terán-Cerqueda, García-Stivalet, Limón-Flores, Armenta-Castro, Rodríguez-Martínez, Leon-Chavez, Vallejo-Ruiz, Hassane, Pérez-Tapia, Ortiz-Navarrete, Guzman and Pelayo.)- Published
- 2021
- Full Text
- View/download PDF
35. Comparison of Multiple Clinical Testing Modalities for Assessment of NPM1-Mutant AML.
- Author
-
Lopez A, Patel S, Geyer JT, Racchumi J, Chadburn A, Simonson P, Ouseph MM, Inghirami G, Mencia-Trinchant N, Guzman ML, Gomez-Arteaga A, Lee S, Desai P, Ritchie EK, Roboz GJ, Tam W, and Kluk MJ
- Abstract
Background: NPM1 mutation status can influence prognosis and management in AML. Accordingly, clinical testing (i.e., RT-PCR, NGS and IHC) for mutant NPM1 is increasing in order to detect residual disease in AML, alongside flow cytometry (FC). However, the relationship of the results from RT-PCR to traditional NGS, IHC and FC is not widely known among many practitioners. Herein, we aim to: i) describe the performance of RT-PCR compared to traditional NGS and IHC for the detection of mutant NPM1 in clinical practice, and also compare it to FC, and ii) provide our observations regarding the advantages and disadvantages of each approach in order to inform future clinical testing algorithms., Methods: Peripheral blood and bone marrow samples collected for clinical testing at variable time points during patient management were tested by quantitative, real-time, RT-PCR and results were compared to findings from a Myeloid NGS panel, mutant NPM1 IHC and FC., Results: RT-PCR showed superior sensitivity compared to NGS, IHC and FC with the main challenge of NGS, IHC and FC being the ability to identify a low disease burden (<0.5% NCN by RT-PCR). Nevertheless, the positive predictive value of NGS, IHC and FC were each ≥ 80% indicating that positive results by those assays are typically associated with RT-PCR positivity. IHC, unlike bulk methods (RT-PCR, NGS and FC), is able provide information regarding cellular/architectural context of disease in biopsies. FC did not identify any NPM1 -mutated residual disease not already detected by RT-PCR, NGS or IHC., Conclusion: Overall, our findings demonstrate that RT-PCR shows superior sensitivity compared to a traditional Myeloid NGS, suggesting the need for "deep-sequencing" NGS panels for NGS-based monitoring of residual disease in NPM1 -mutant AML. IHC provides complementary cytomorphologic information to RT-PCR. Lastly, FC may not be necessary in the setting of post-therapy follow up for NPM1 -mutated AML. Together, these findings can help inform future clinical testing algorithms., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Lopez, Patel, Geyer, Racchumi, Chadburn, Simonson, Ouseph, Inghirami, Mencia-Trinchant, Guzman, Gomez-Arteaga, Lee, Desai, Ritchie, Roboz, Tam and Kluk.)
- Published
- 2021
- Full Text
- View/download PDF
36. Small-molecule inhibitors targeting Polycomb repressive complex 1 RING domain.
- Author
-
Shukla S, Ying W, Gray F, Yao Y, Simes ML, Zhao Q, Miao H, Cho HJ, González-Alonso P, Winkler A, Lund G, Purohit T, Kim E, Zhang X, Ray JM, He S, Nikolaidis C, Ndoj J, Wang J, Jaremko Ł, Jaremko M, Ryan RJH, Guzman ML, Grembecka J, and Cierpicki T
- Subjects
- Cell Differentiation drug effects, Dose-Response Relationship, Drug, Humans, K562 Cells, Models, Molecular, Molecular Structure, Polycomb Repressive Complex 1 genetics, Polycomb Repressive Complex 1 metabolism, Small Molecule Libraries chemical synthesis, Small Molecule Libraries chemistry, Ubiquitination drug effects, Polycomb Repressive Complex 1 antagonists & inhibitors, Small Molecule Libraries pharmacology
- Abstract
Polycomb repressive complex 1 (PRC1) is an essential chromatin-modifying complex that monoubiquitinates histone H2A and is involved in maintaining the repressed chromatin state. Emerging evidence suggests PRC1 activity in various cancers, rationalizing the need for small-molecule inhibitors with well-defined mechanisms of action. Here, we describe the development of compounds that directly bind to RING1B-BMI1, the heterodimeric complex constituting the E3 ligase activity of PRC1. These compounds block the association of RING1B-BMI1 with chromatin and inhibit H2A ubiquitination. Structural studies demonstrate that these inhibitors bind to RING1B by inducing the formation of a hydrophobic pocket in the RING domain. Our PRC1 inhibitor, RB-3, decreases the global level of H2A ubiquitination and induces differentiation in leukemia cell lines and primary acute myeloid leukemia (AML) samples. In summary, we demonstrate that targeting the PRC1 RING domain with small molecules is feasible, and RB-3 represents a valuable chemical tool to study PRC1 biology.
- Published
- 2021
- Full Text
- View/download PDF
37. Macrophages in Acute Myeloid Leukaemia: Significant Players in Therapy Resistance and Patient Outcomes.
- Author
-
Miari KE, Guzman ML, Wheadon H, and Williams MTS
- Abstract
Acute Myeloid Leukaemia (AML) is a commonly occurring severe haematological malignancy, with most patients exhibiting sub-optimal clinical outcomes. Therapy resistance significantly contributes towards failure of traditional and targeted treatments, disease relapse and mortality in AML patients. The mechanisms driving therapy resistance in AML are not fully understood, and approaches to overcome therapy resistance are important for curative therapies. To date, most studies have focused on therapy resistant mechanisms inherent to leukaemic cells (e.g., TP53 mutations), overlooking to some extent, acquired mechanisms of resistance through extrinsic processes. In the bone marrow microenvironment (BMME), leukaemic cells interact with the surrounding bone resident cells, driving acquired therapy resistance in AML. Growing evidence suggests that macrophages, highly plastic immune cells present in the BMME, play a role in the pathophysiology of AML. Leukaemia-supporting macrophage subsets (CD163
+ CD206+ ) are elevated in preclinical in vivo models of AML and AML patients. However, the relationship between macrophages and therapy resistance in AML warrants further investigation. In this review, we correlate the potential links between macrophages, the development of therapy resistance, and patient outcomes in AML. We specifically focus on macrophage reprogramming by AML cells, macrophage-driven activation of anti-cell death pathways in AML cells, and the association between macrophage phenotypes and clinical outcomes in AML, including their potential prognostic value. Lastly, we discuss therapeutic targeting of macrophages, as a strategy to circumvent therapy resistance in AML, and discuss how emerging genomic and proteomic-based approaches can be utilised to address existing challenges in this research field., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Miari, Guzman, Wheadon and Williams.)- Published
- 2021
- Full Text
- View/download PDF
38. Sustained dual release of ciprofloxacin and lidocaine from ionic exchange responding film based on alginate and hyaluronate for wound healing.
- Author
-
Sanchez MF, Guzman ML, Apas AL, Alovero FDL, and Olivera ME
- Subjects
- Anti-Bacterial Agents therapeutic use, Lidocaine, Staphylococcus aureus, Wound Healing, Alginates, Ciprofloxacin
- Abstract
This study presents a new antibiotic-anesthetic film (AA-film) based on natural polyelectrolytes ionically complexed with lidocaine and ciprofloxacin to manage pain associated with infected wounds. The rational selection of the components resulted in the AA-films being transparent, compatible with wound skin pH and highly water vapor permeable. The drug release properties evaluated in saline solution and water revealed an ionic exchange mechanism for the release of both drugs and showed that ciprofloxacin acts as a cross-linker, as was confirmed by rheological evaluation. The in vitro antimicrobial efficacy against S. aureus and P. aeruginosa was demonstrated. Furthermore, AA-films exhibit a high fluid absorption capacity and act as a physical barrier for microorganisms. This work highlights the great potential of this smart system as an attractive dressing for skin wounds, surpassing currently available treatments., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
39. Efficacy of topical risedronate and risedronate - Eudragit E complex in a model of cutaneous leishmaniasis induced by Leishmania (Leishmania) amazonensis .
- Author
-
Peralta MF, Guzman ML, Bracamonte ME, Marco JD, Olivera ME, Carrer DC, and Barroso PA
- Abstract
An efficacious topical treatment for cutaneous leishmaniasis (CL) is highly desirable but still an ongoing challenge. Systemic risedronate (Ris) has been reported to have anti-leishmanial properties and Eudragit EPO (EuE) has shown in vitro activity against L. (L.) amazonensis . The aim of this work was to investigate the in vivo efficacy of topical Ris and EuE-Ris complexes on CL. Surface charge and Ris release kinetics from the different dispersions were analyzed. BALB/c mice were infected intradermally with promastigotes of L. (L.) amazonensis . Ulcers were treated with Ris or EuE-Ris hydrogels. All the lesions that received topical Ris or EuE-Ris showed an improvement with respect to control: reduction of ulcer average size, cicatrization, flattened edges and no signs of necrosis. In addition, a marked parasitic inhibition of 69.5 and 73.7% was observed in the groups treated with Ris and EuE-Ris, respectively, with the IgG2a levels indicating a tendency towards cure. The results are promising and the system should now be enhanced to achieve total parasite elimination., Competing Interests: The authors declare no conflict of interest., (© 2021 Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
40. Targeting the epichaperome as an effective precision medicine approach in a novel PML-SYK fusion acute myeloid leukemia.
- Author
-
Sugita M, Wilkes DC, Bareja R, Eng KW, Nataraj S, Jimenez-Flores RA, Yan L, De Leon JP, Croyle JA, Kaner J, Merugu S, Sharma S, MacDonald TY, Noorzad Z, Panchal P, Pancirer D, Cheng S, Xiang JZ, Olson L, Van Besien K, Rickman DS, Mathew S, Tam W, Rubin MA, Beltran H, Sboner A, Hassane DC, Chiosis G, Elemento O, Roboz GJ, Mosquera JM, and Guzman ML
- Abstract
The epichaperome is a new cancer target composed of hyperconnected networks of chaperome members that facilitate cell survival. Cancers with an altered chaperone configuration may be susceptible to epichaperome inhibitors. We developed a flow cytometry-based assay for evaluation and monitoring of epichaperome abundance at the single cell level, with the goal of prospectively identifying patients likely to respond to epichaperome inhibitors, to measure target engagement, and dependency during treatment. As proof of principle, we describe a patient with an unclassified myeloproliferative neoplasm harboring a novel PML-SYK fusion, who progressed to acute myeloid leukemia despite chemotherapy and allogeneic stem cell transplant. The leukemia was identified as having high epichaperome abundance. We obtained compassionate access to an investigational epichaperome inhibitor, PU-H71. After 16 doses, the patient achieved durable complete remission. These encouraging results suggest that further investigation of epichaperome inhibitors in patients with abundant baseline epichaperome levels is warranted.
- Published
- 2021
- Full Text
- View/download PDF
41. Inhibiting autophagy targets human leukemic stem cells and hypoxic AML blasts by disrupting mitochondrial homeostasis.
- Author
-
Dykstra KM, Fay HRS, Massey AC, Yang N, Johnson M, Portwood S, Guzman ML, and Wang ES
- Subjects
- Animals, Autophagy, Homeostasis, Humans, Hypoxia, Mice, Mitochondria, Stem Cells, Tumor Microenvironment, Leukemia, Myeloid, Acute drug therapy
- Abstract
Leukemia stem cells (LSCs) and therapy-resistant acute myeloid leukemia (AML) blasts contribute to the reinitiation of leukemia after remission, necessitating therapeutic interventions that target these populations. Autophagy is a prosurvival process that allows for cells to adapt to a variety of stressors. Blocking autophagy pharmacologically by using mechanistically distinct inhibitors induced apoptosis and prevented colony formation in primary human AML cells. The most effective inhibitor, bafilomycin A1 (Baf A1), also prevented the in vivo maintenance of AML LSCs in NSG mice. To understand why Baf A1 exerted the most dramatic effects on LSC survival, we evaluated mitochondrial function. Baf A1 reduced mitochondrial respiration and stabilized PTEN-induced kinase-1 (PINK-1), which initiates autophagy of mitochondria (mitophagy). Interestingly, with the autophagy inhibitor chloroquine, levels of enhanced cell death and reduced mitochondrial respiration phenocopied the effects of Baf A1 only when cultured in hypoxic conditions that mimic the marrow microenvironment (1% O2). This indicates that increased efficacy of autophagy inhibitors in inducing AML cell death can be achieved by concurrently inducing mitochondrial damage and mitophagy (pharmacologically or by hypoxic induction) and blocking mitochondrial degradation. In addition, prolonged exposure of AML cells to hypoxia induced autophagic flux and reduced chemosensitivity to cytarabine (Ara-C), which was reversed by autophagy inhibition. The combination of Ara-C with Baf A1 also decreased tumor burden in vivo. These findings demonstrate that autophagy is critical for mitochondrial homeostasis and survival of AML cells in hypoxia and support the development of autophagy inhibitors as novel therapeutic agents for AML., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
42. BCL6 maintains survival and self-renewal of primary human acute myeloid leukemia cells.
- Author
-
Kawabata KC, Zong H, Meydan C, Wyman S, Wouters BJ, Sugita M, Goswami S, Albert M, Yip W, Roboz GJ, Chen Z, Delwel R, Carroll M, Mason CE, Melnick A, and Guzman ML
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis, Cell Self Renewal, Cytarabine therapeutic use, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Gene Knockdown Techniques, Hematopoietic Stem Cells cytology, Humans, Indoles pharmacology, Indoles therapeutic use, Leukemia, Myeloid, Acute genetics, Mice, Mice, Inbred NOD, Mice, SCID, Neoplastic Stem Cells cytology, Proto-Oncogene Mas, Proto-Oncogene Proteins c-bcl-6 antagonists & inhibitors, RNA Interference, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Neoplasm biosynthesis, RNA, Neoplasm genetics, RNA, Small Interfering genetics, RNA, Small Interfering pharmacology, RNA-Seq, Radiation Chimera, Thiazolidinediones pharmacology, Thiazolidinediones therapeutic use, Tumor Stem Cell Assay, Xenograft Model Antitumor Assays, Leukemia, Myeloid, Acute pathology, Neoplasm Proteins physiology, Proto-Oncogene Proteins c-bcl-6 physiology
- Abstract
B-cell lymphoma 6 (BCL6) is a transcription repressor and proto-oncogene that plays a crucial role in the innate and adaptive immune system and lymphoid neoplasms. However, its role in myeloid malignancies remains unclear. Here, we explored the role of BCL6 in acute myeloid leukemia (AML). BCL6 was expressed at variable and often high levels in AML cell lines and primary AML samples. AMLs with higher levels of BCL6 were generally sensitive to treatment with BCL6 inhibitors, with the exception of those with monocytic differentiation. Gene expression profiling of AML cells treated with a BCL6 inhibitor revealed induction of BCL6-repressed target genes and transcriptional programs linked to DNA damage checkpoints and downregulation of stem cell genes. Ex vivo treatment of primary AML cells with BCL6 inhibitors induced apoptosis and decreased colony-forming capacity, which correlated with the levels of BCL6 expression. Importantly, inhibition or knockdown of BCL6 in primary AML cells resulted in a significant reduction of leukemia-initiating capacity in mice, suggesting ablation of leukemia repopulating cell functionality. In contrast, BCL6 knockout or inhibition did not suppress the function of normal hematopoietic stem cells. Treatment with cytarabine further induced BCL6 expression, and the levels of BCL6 induction were correlated with resistance to cytarabine. Treatment of AML patient-derived xenografts with BCL6 inhibitor plus cytarabine suggested enhanced antileukemia activity with this combination. Hence, pharmacologic inhibition of BCL6 might provide a novel therapeutic strategy for ablation of leukemia-repopulating cells and increased responsiveness to chemotherapy., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
43. Clonal hematopoiesis before, during, and after human spaceflight.
- Author
-
Mencia-Trinchant N, MacKay MJ, Chin C, Afshinnekoo E, Foox J, Meydan C, Butler D, Mozsary C, Vernice NA, Darby C, Schatz MC, Bailey SM, Melnick AM, Guzman ML, Bolton K, Braunstein LZ, Garrett-Bakelman F, Levine RL, Hassane DC, and Mason CE
- Published
- 2021
- Full Text
- View/download PDF
44. Clonal Hematopoiesis Before, During, and After Human Spaceflight.
- Author
-
Mencia-Trinchant N, MacKay MJ, Chin C, Afshinnekoo E, Foox J, Meydan C, Butler D, Mozsary C, Vernice NA, Darby C, Schatz MC, Bailey SM, Melnick AM, Guzman ML, Bolton K, Braunstein LZ, Garrett-Bakelman F, Levine RL, Hassane DC, and Mason CE
- Subjects
- Adult, Astronauts, Clone Cells, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, DNA-Binding Proteins genetics, Dioxygenases genetics, Female, Hematologic Neoplasms genetics, Hematopoiesis genetics, Hematopoiesis physiology, Humans, Male, Middle Aged, Mutation genetics, Neoplasms genetics, Risk Factors, Space Flight, Time Factors, Clonal Hematopoiesis genetics, Clonal Hematopoiesis physiology, Weightlessness adverse effects
- Abstract
Clonal hematopoiesis (CH) occurs when blood cells harboring an advantageous mutation propagate faster than others. These mutations confer a risk for hematological cancers and cardiovascular disease. Here, we analyze CH in blood samples from a pair of twin astronauts over 4 years in bulk and fractionated cell populations using a targeted CH panel, linked-read whole-genome sequencing, and deep RNA sequencing. We show CH with distinct mutational profiles and increasing allelic fraction that includes a high-risk, TET2 clone in one subject and two DNMT3A mutations on distinct alleles in the other twin. These astronauts exhibit CH almost two decades prior to the mean age at which it is typically detected and show larger shifts in clone size than age-matched controls or radiotherapy patients, based on a longitudinal cohort of 157 cancer patients. As such, longitudinal monitoring of CH may serve as an important metric for overall cancer and cardiovascular risk in astronauts., Competing Interests: Declaration of Interests No relevant conflicts apply to this work, but C.E.M. is a cofounder of Onegevity and Biotia. D.H. and C.E.M. also have positions at Tempus Labs., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
45. Synthesis, Crystallography, and Anti-Leukemic Activity of the Amino Adducts of Dehydroleucodine.
- Author
-
Ordóñez PE, Mery DE, Sharma KK, Nemu S, Reynolds WF, Enriquez RG, Burns DC, Malagón O, Jones DE, Guzman ML, and Compadre CM
- Subjects
- Cell Line, Tumor, Crystallography, X-Ray, Gene Expression Regulation, Leukemic drug effects, HSP70 Heat-Shock Proteins genetics, Heme Oxygenase-1 genetics, Humans, Lactones chemical synthesis, Lactones pharmacology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Morpholines chemistry, NF-kappa B p50 Subunit genetics, Piperidines chemistry, Sesquiterpenes chemical synthesis, Sesquiterpenes pharmacology, Tyramine chemistry, Cell Proliferation drug effects, Lactones chemistry, Leukemia, Myeloid, Acute drug therapy, Leukocytes, Mononuclear drug effects, Sesquiterpenes chemistry
- Abstract
Dehydroleucodine is a bioactive sesquiterpene lactone. Herein, four dehydroleucodine amino derivatives were synthesized using the amines proline, piperidine, morpholine, and tyramine, and spectroscopic methods and single-crystal X-ray diffraction unambiguously established their structures. The cytotoxic activity of these compounds was evaluated against eight acute myeloid leukemia cell lines, and their toxicity to peripheral blood mononuclear cells was also determined. The proline adduct was the most active compound, it showed anti-leukemic activity, upregulated heme oxygenase 1 (HMOX1) and the primary stress-inducible isoform of the heath shock 70 kDa protein 1 (HSPA1A), and downregulated NFkB1 transcription, it was also found to be about 270 times more water soluble than dehydroleucodine.
- Published
- 2020
- Full Text
- View/download PDF
46. CD123 as a Therapeutic Target Against Malignant Stem Cells.
- Author
-
Sugita M and Guzman ML
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Combined Modality Therapy, Gene Expression, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Humans, Molecular Targeted Therapy, Neoplasms metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Biomarkers, Tumor, Interleukin-3 Receptor alpha Subunit antagonists & inhibitors, Neoplasms drug therapy, Neoplasms etiology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism
- Abstract
Hematopoiesis is a tightly regulated process that originates from highly specialized cells, hematopoietic stem cells (HSCs). Many cancers can arise and be maintained by malignant stem cells. In acute myeloid leukemia, leukemic stem cells (LSCs) are identified by their immunophenotype, which is partly shared with normal HSCs (CD34
+ CD38- ). However, LSCs also possess unique immunophenotypic features that can be used to distinguish them from HSCs and therapeutically target them. One such unique immunophenotypic marker is CD123, found to be aberrantly expressed in leukemic stem, progenitor, and blast cells. Thus, CD123 is sought as an attractive target to eliminate LSCs., Competing Interests: Disclosure M.L. Guzman reports receiving research funding (R01 CA234478) from NIH. M.L. Guzman receives research funding from Cellectis and and honoraria from SeqRx. M. Sugita has nothing to disclose., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
47. Clonal Hematopoiesis and Premalignant Diseases.
- Author
-
Kaner J, Desai P, Mencia-Trinchant N, Guzman ML, Roboz GJ, and Hassane DC
- Subjects
- Animals, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Hematologic Neoplasms therapy, Hematopoietic Stem Cells metabolism, Humans, Leukemia genetics, Leukemia metabolism, Leukemia therapy, Mutation, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Precancerous Conditions genetics, Precancerous Conditions metabolism, Clonal Hematopoiesis genetics, Hematologic Neoplasms pathology, Hematopoietic Stem Cells pathology, Leukemia pathology, Precancerous Conditions pathology
- Abstract
Clonal hematopoiesis (CH) arises when mutations in the hematopoietic system confer a fitness advantage to specific clones, thereby favoring their disproportionate growth. The presence of CH increases with age and environmental exposures such as cytotoxic chemotherapy or radiotherapy. The most frequent mutations occur in epigenetic regulators, such as DNMT3A , TET2 , and ASXL1 , leading to dysregulation of tumor suppressor function, pathogen response, and inflammation. These dysregulated processes elevate risk of overall mortality, cardiovascular disease, and eventual hematologic malignancy (HM). CH is likely acting as an initiating event leading to HM when followed by cooperating mutations. However, further evidence suggests that CH exerts a bystander influence through its pro-inflammatory properties. Delineating the mechanisms that lead to the onset and expansion of CH as well as its contribution to risk of HM is crucial to defining a management and intervention strategy. In this review, we discuss the potential causes, consequences, technical considerations, and possible management strategies for CH in the context of HMs and pre-HMs., (Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. A Chemical Biology Approach to the Chaperome in Cancer-HSP90 and Beyond.
- Author
-
Taldone T, Wang T, Rodina A, Pillarsetty NVK, Digwal CS, Sharma S, Yan P, Joshi S, Pagare PP, Bolaender A, Roboz GJ, Guzman ML, and Chiosis G
- Subjects
- Animals, Antineoplastic Agents pharmacology, Biology, Decision Making, Drug Design, HEK293 Cells, Humans, K562 Cells, Kinetics, Mice, NIH 3T3 Cells, Neoplasms drug therapy, Protein Binding, Gene Expression Regulation, Neoplastic, HSP70 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism, Molecular Chaperones metabolism, Neoplasms metabolism
- Abstract
Cancer is often associated with alterations in the chaperome, a collection of chaperones, cochaperones, and other cofactors. Changes in the expression levels of components of the chaperome, in the interaction strength among chaperome components, alterations in chaperome constituency, and in the cellular location of chaperome members, are all hallmarks of cancer. Here we aim to provide an overview on how chemical biology has played a role in deciphering such complexity in the biology of the chaperome in cancer and in other diseases. The focus here is narrow and on pathologic changes in the chaperome executed by enhancing the interaction strength between components of distinct chaperome pathways, specifically between those of HSP90 and HSP70 pathways. We will review chemical tools and chemical probe-based assays, with a focus on HSP90. We will discuss how kinetic binding, not classical equilibrium binding, is most appropriate in the development of drugs and probes for the chaperome in disease. We will then present our view on how chaperome inhibitors may become potential drugs and diagnostics in cancer., (Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
49. The therapeutic landscape for cells engineered with chimeric antigen receptors.
- Author
-
MacKay M, Afshinnekoo E, Rub J, Hassan C, Khunte M, Baskaran N, Owens B, Liu L, Roboz GJ, Guzman ML, Melnick AM, Wu S, and Mason CE
- Subjects
- Cell- and Tissue-Based Therapy, Clinical Trials as Topic, Gene Expression Regulation, Neoplastic, Humans, Neoplasms genetics, Neoplasms metabolism, Cell Engineering methods, Receptors, Chimeric Antigen metabolism
- Abstract
Despite the global rapid increase in the number of clinical trials employing chimeric antigen receptors (CARs), no comprehensive survey of their scope, targets and design exists. In this study, we present an interactive CAR clinical trial database, spanning 64 targets deployed in T cells (CAR-T), natural killer cells (CAR-NK) or mixtures (CAR-NK/T) from over 500 clinical trials in 20 countries, encompassing >20,000 patients. By combining these data with transcriptional and proteomic data, we create a 'targetable landscape' for CAR cell therapies based on 13,206 proteins and RNAs across 78 tissues, 124 cell types and 20 cancer types. These data suggest a landscape of over 100 single targets and over 100,000 target pairs using logical switches for CAR cell engineering. Our analysis of the CAR cellular therapeutic landscape may aid the design of future therapies, improve target-to-patient matching, and ultimately help inform our understanding of CAR therapy's safety and efficacy.
- Published
- 2020
- Full Text
- View/download PDF
50. Chemical probes and methods for single-cell detection and quantification of epichaperomes in hematologic malignancies.
- Author
-
Merugu S, Sharma S, Kaner J, Digwal C, Sugita M, Joshi S, Taldone T, Guzman ML, and Chiosis G
- Subjects
- Flow Cytometry, Humans, Hematologic Neoplasms
- Abstract
Detection of protein connectivity dysfunctions in biological samples, i.e., informing on how protein-protein interactions change from a normal to a disease state, is important for both biomedical research and clinical development. The epichaperome is an executor of protein connectivity dysfunction in disease, and thus a surrogate for its detection. This chapter will detail on published methods for epichaperome detection and quantification that combine the advantages of multiparameter flow cytometry with those of the PU-FITC fluorescently labeled epichaperome detection probe. It will offer a comprehensive method description that includes the synthesis and characterization of an epichaperome detection probe and of the negative control probe, the preparation of the biospecimen for epichaperome analysis, the execution of the epichaperome detection and quantification assay and lastly, the data acquisition and analysis. The method provides, at single-cell level, the functional signature of cells, differentiating itself from other single-cell methods that provide a catalog of molecules., Competing Interests: Declaration of interests Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College hold the intellectual rights to PU-FITC. Samus Therapeutics, of which G.C. has partial ownership and is a member of its board of directors, has licensed PU-FITC. G.C., M.L.G., and T.T. are inventors on the licensed intellectual property. All other authors declare no competing interests., (© 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.