326 results on '"Haber DA"'
Search Results
2. Abstract P2-01-14: circulating tumor cells in breast cancer exhibit dynamic changes in epithelial and mesenchymal cell composition
- Author
-
Yu, M, primary, Bardia, A, additional, Wittner, BS, additional, Stott, SL, additional, Smas, ME, additional, Ting, DT, additional, Isakoff, SJ, additional, Ciciliano, JC, additional, Wells, MN, additional, Shah, AM, additional, Concannon, KF, additional, Sequist, LV, additional, Brachtel, E, additional, Sgroi, D, additional, Baselga, J, additional, Ramaswamy, S, additional, Toner, M, additional, Haber, DA, additional, and Maheswaran, S, additional
- Published
- 2012
- Full Text
- View/download PDF
3. Multidrug resistance (MDR 1) in leukemia: is it time to test? [editorial] [see comments]
- Author
-
Haber, DA, primary
- Published
- 1992
- Full Text
- View/download PDF
4. First-line gefitinib in patients with advanced non--small-cell lung cancer harboring somatic EGFR mutations.
- Author
-
Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Jänne PA, Joshi VA, McCollum D, Evans TL, Muzikansky A, Kuhlmann GL, Han M, Goldberg JS, Settleman J, Iafrate AJ, Engelman JA, Haber DA, Johnson BE, and Lynch TJ
- Published
- 2008
5. Case 28-2003: a 51-year-old premenopausal woman with newly diagnosed breast cancer and a strong family history of breast cancer.
- Author
-
Ryan PD, Haber DA, Shannon KM, Smith BL, and Fan MJ
- Published
- 2003
6. Yeast, flies, worms, and fish in the study of human disease.
- Author
-
Hariharan IK and Haber DA
- Subjects
- *
BIOLOGICAL models , *DISEASES , *FISHES , *GENETIC techniques , *HELMINTHS , *HUMAN genome , *INSECTS , *MEDICAL genetics , *GENETIC mutation , *RESEARCH , *YEAST , *GENETIC testing - Published
- 2003
7. Pulmonary adenocarcinomas with mutant epidermal growth factor receptors.
- Author
-
Riemenschneider MJ, Bell DW, Haber DA, and Louis DN
- Published
- 2005
8. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.
- Author
-
Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, and Haber DA
- Published
- 2004
9. Germ-line BRCA1 mutations in Jewish and non-Jewish women with early-onset breast cancer.
- Author
-
FitzGerald MG, MacDonald DJ, Krainer M, Hoover I, O'Neil E, Unsal H, Silva-Arrieto S, Finkelstein DM, Beer-Romero P, Englert C, Sgroi DC, Smith BL, Younger JW, Garber JE, Duda RB, Mayzel KA, Isselbacher KJ, Friend SH, and Haber DA
- Published
- 1996
10. EGFR mutations and sensitivity to gefitinib.
- Author
-
Sorscher SM, Rich JN, Rasheed BKA, Yan H, Rossi G, Marchioni A, Longo L, Haber DA, Bell DW, and Lynch TJ
- Published
- 2004
11. Combination Diagnostics: Adding Blood-Based ctDNA Screening to Low-Dose CT Imaging for Early Detection of Lung Cancer.
- Author
-
Haber DA and Skates SJ
- Subjects
- Humans, Biomarkers, Tumor, Lung Neoplasms genetics, Lung Neoplasms diagnosis, Lung Neoplasms diagnostic imaging, Circulating Tumor DNA blood, Early Detection of Cancer methods, Tomography, X-Ray Computed methods
- Abstract
Annual low-dose CT screening of individuals with a smoking history identifies early curable lung tumors and reduces cancer mortality by 20%, yet only a minority of eligible patients undergo such monitoring. Mazzone and colleagues apply a blood-based cfDNA fragmentomic assay as a high-sensitivity/low-specificity pre-screen to help stratify individuals who may benefit most from more definitive low-dose CT imaging. See related article by Mazzone et al., p. 2224., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
12. Developmental mosaicism underlying EGFR-mutant lung cancer presenting with multiple primary tumors.
- Author
-
Burr R, Leshchiner I, Costantino CL, Blohmer M, Sundaresan T, Cha J, Seeger K, Guay S, Danysh BP, Gore I, Jacobs RA, Slowik K, Utro F, Rhrissorrakrai K, Levovitz C, Barth JL, Dubash T, Chirn B, Parida L, Sequist LV, Lennerz JK, Mino-Kenudson M, Maheswaran S, Naxerova K, Getz G, and Haber DA
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Mutation, Exome Sequencing, Genetic Predisposition to Disease, Germ-Line Mutation, Mosaicism, ErbB Receptors genetics, Lung Neoplasms genetics, Neoplasms, Multiple Primary genetics, Carcinoma, Non-Small-Cell Lung genetics
- Abstract
Although the development of multiple primary tumors in smokers with lung cancer can be attributed to carcinogen-induced field cancerization, the occurrence of multiple tumors at presentation in individuals with EGFR-mutant lung cancer who lack known environmental exposures remains unexplained. In the present study, we identified ten patients with early stage, resectable, non-small cell lung cancer who presented with multiple, anatomically distinct, EGFR-mutant tumors. We analyzed the phylogenetic relationships among multiple tumors from each patient using whole-exome sequencing (WES) and hypermutable poly(guanine) (poly(G)) repeat genotyping as orthogonal methods for lineage tracing. In four patients, developmental mosaicism, assessed by WES and poly(G) lineage tracing, indicates a common non-germline cell of origin. In two other patients, we identified germline EGFR variants, which confer moderately enhanced signaling when modeled in vitro. Thus, in addition to germline variants, developmental mosaicism defines a distinct mechanism of genetic predisposition to multiple EGFR-mutant primary tumors, with implications for their etiology and clinical management., Competing Interests: Competing interests: L.V.S. has institutional research funding from AstraZeneca, Novartis and Delfi Diagnostics and has received consulting fees from AstraZeneca, Janssen, Pfizer and Genentech. I.L. serves as a consultant for PACT Pharma Inc., has stock, is on the board and serves as a consultant for ennov1 LLC, and is on the board and holds equity in Nord Bio, Inc. G.G. receives research funds from IBM, Pharmacyclics/Abbvie, Bayer, Genentech and Ultima Genomics, and is also an inventor on patent applications filed by the Broad Institute related to MSMuTect, MSMutSig, POLYSOLVER, SignatureAnalyzer-GPU, MSIDetect and MinumuMM-seq. He is a founder and consultant and holds privately held equity in Scorpion Therapeutics; he is also a founder of, and holds privately held equity in, PreDICTA Biosciences. M.M.-K. has served as a compensated consultant for AstraZeneca, Pfizer, Repare, Sanofi, AbbVie and Daiichi Sankyo and has received royalties from Elsevier. The remaining authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Correction: Landscape of Targeted Anti-Cancer Drug Synergies in Melanoma Identifies a Novel BRAF-VEGFR/PDGFR Combination Treatment.
- Author
-
Friedman AA, Amzallag A, Pruteanu-Malinici I, Baniya S, Cooper ZA, Piris A, Hargreaves L, Igras V, Frederick DT, Lawrence DP, Haber DA, Flaherty KT, Wargo JA, Ramaswamy S, Benes CH, and Fisher DE
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0140310.]., (Copyright: © 2024 Friedman et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
14. Targeting mosquito X-chromosomes reveals complex transmission dynamics of sex ratio distorting gene drives.
- Author
-
Haber DA, Arien Y, Lamdan LB, Alcalay Y, Zecharia C, Krsticevic F, Yonah ES, Avraham RD, Krzywinska E, Krzywinski J, Marois E, Windbichler N, and Papathanos PA
- Subjects
- Animals, Male, Female, Mosquito Vectors genetics, Genes, X-Linked, CRISPR-Cas Systems, Spermatozoa metabolism, Animals, Genetically Modified, Sex Ratio, Anopheles genetics, X Chromosome genetics, Drosophila melanogaster genetics, Gene Drive Technology methods, Spermatogenesis genetics
- Abstract
Engineered sex ratio distorters (SRDs) have been proposed as a powerful component of genetic control strategies designed to suppress harmful insect pests. Two types of CRISPR-based SRD mechanisms have been proposed: X-shredding, which eliminates X-bearing sperm, and X-poisoning, which eliminates females inheriting disrupted X-chromosomes. These differences can have a profound impact on the population dynamics of SRDs when linked to the Y-chromosome: an X-shredder is invasive, constituting a classical meiotic Y-drive, whereas X-poisoning is self-limiting, unable to invade but also insulated from selection. Here, we establish X-poisoning strains in the malaria vector Anopheles gambiae targeting three X-linked genes during spermatogenesis, resulting in male bias. We find that sex distortion is primarily driven by a loss of X-bearing sperm, with limited evidence for postzygotic lethality of female progeny. By leveraging a Drosophila melanogaster model, we show unambiguously that engineered SRD traits can operate differently in these two insects. Unlike X-shredding, X-poisoning could theoretically operate at early stages of spermatogenesis. We therefore explore premeiotic Cas9 expression to target the mosquito X-chromosome. We find that, by pre-empting the onset of meiotic sex chromosome inactivation, this approach may enable the development of Y-linked SRDs if mutagenesis of spermatogenesis-essential genes is functionally balanced., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. DrugMap: A quantitative pan-cancer analysis of cysteine ligandability.
- Author
-
Takahashi M, Chong HB, Zhang S, Yang TY, Lazarov MJ, Harry S, Maynard M, Hilbert B, White RD, Murrey HE, Tsou CC, Vordermark K, Assaad J, Gohar M, Dürr BR, Richter M, Patel H, Kryukov G, Brooijmans N, Alghali ASO, Rubio K, Villanueva A, Zhang J, Ge M, Makram F, Griesshaber H, Harrison D, Koglin AS, Ojeda S, Karakyriakou B, Healy A, Popoola G, Rachmin I, Khandelwal N, Neil JR, Tien PC, Chen N, Hosp T, van den Ouweland S, Hara T, Bussema L, Dong R, Shi L, Rasmussen MQ, Domingues AC, Lawless A, Fang J, Yoda S, Nguyen LP, Reeves SM, Wakefield FN, Acker A, Clark SE, Dubash T, Kastanos J, Oh E, Fisher DE, Maheswaran S, Haber DA, Boland GM, Sade-Feldman M, Jenkins RW, Hata AN, Bardeesy NM, Suvà ML, Martin BR, Liau BB, Ott CJ, Rivera MN, Lawrence MS, and Bar-Peled L
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Ligands, Melanoma metabolism, NF-kappa B chemistry, NF-kappa B metabolism, Oxidation-Reduction, Signal Transduction, SOXE Transcription Factors chemistry, SOXE Transcription Factors metabolism, Cysteine metabolism, Cysteine chemistry, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
Cysteine-focused chemical proteomic platforms have accelerated the clinical development of covalent inhibitors for a wide range of targets in cancer. However, how different oncogenic contexts influence cysteine targeting remains unknown. To address this question, we have developed "DrugMap," an atlas of cysteine ligandability compiled across 416 cancer cell lines. We unexpectedly find that cysteine ligandability varies across cancer cell lines, and we attribute this to differences in cellular redox states, protein conformational changes, and genetic mutations. Leveraging these findings, we identify actionable cysteines in NF-κB1 and SOX10 and develop corresponding covalent ligands that block the activity of these transcription factors. We demonstrate that the NF-κB1 probe blocks DNA binding, whereas the SOX10 ligand increases SOX10-SOX10 interactions and disrupts melanoma transcriptional signaling. Our findings reveal heterogeneity in cysteine ligandability across cancers, pinpoint cell-intrinsic features driving cysteine targeting, and illustrate the use of covalent probes to disrupt oncogenic transcription-factor activity., Competing Interests: Declaration of interests D.E.F. has a financial interest in Soltego. S.M. and D.A.H. are cofounders of TellBio. G.M.B. has sponsored research agreements with Olink Proteomics, Teiko Bio, InterVenn Biosciences, and Palleon Pharmaceuticals; served on advisory boards for Iovance, Merck, Nektar Therapeutics, Novartis, and Ankyra Therapeutics; consults for Merck, InterVenn Biosciences, Iovance, and Ankyra Therapeutics; and holds equity in Ankyra Therapeutics. M.S.-F. received funding from Calico Life Sciences, Bristol-Myers Squibb, Istari Oncology, and consultants for Galvanize Therapeutics. R.W.J. is a member of the advisory board for/has a financial interest in Xsphera Biosciences Inc. R.W.J. has received honoraria from Incyte (invited speaker), G1 Therapeutics (advisory board), and Bioxcel Therapeutics (invited speaker). R.W.J. has ownership interest in U.S. patents US20200399573A9 and US20210363595A1. A.N.H. has received grants/research support from Amgen, Blueprint Medicines, BridgeBio, Bristol-Myers Squibb, C4 Therapeutics, Eli Lilly, Novartis, Nuvalent, Pfizer, Roche/Genentech, and Scorpion Therapeutics. A.N.H. has served as a compensated consultant for Amgen, Engine Biosciences, Nuvalent, Oncovalent, Pfizer, TigaTx, and Tolremo Therapeutics. M.L.S. is an equity holder and scientific co-founder/advisory board member of Immunitas Therapeutics. C.J.O. received funding from Scorpion Therapeutics, Gilead Sciences, and eFFECTOR Therapeutics. L.B.-P. is a founder/consultant/holds privately held equity in Scorpion Therapeutics. All relationships for investigators have been reviewed/managed by their respective institutions in accordance with their conflict-of-interest policies. M.M., B.H., R.D.W., H.E.M., C.-C.T., G.K., N.B., J.R.N., and B.R.M. are employees of Scorpion Therapeutics and some hold equity., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product.
- Author
-
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman L, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, and Toner M
- Abstract
Circulating Tumor Cells (CTCs), interrogated by sampling blood from patients with cancer, contain multiple analytes, including intact RNA, high molecular weight DNA, proteins, and metabolic markers. However, the clinical utility of tumor cell-based liquid biopsy has been limited since CTCs are very rare, and current technologies cannot process the blood volumes required to isolate a sufficient number of tumor cells for in-depth assays. We previously described a high-throughput microfluidic prototype utilizing high-flow channels and amplification of cell sorting forces through magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from patients with metastatic cancer, with a median of 2,799 CTCs purified per patient. Isolation of many CTCs from individual patients enables characterization of their morphological and molecular heterogeneity, including cell and nuclear size and RNA expression. It also allows robust detection of gene copy number variation, a definitive cancer marker with potential diagnostic applications. High-volume microfluidic enrichment of CTCs constitutes a new dimension in liquid biopsies.
- Published
- 2024
- Full Text
- View/download PDF
17. Circulating and Imaging Biomarkers of Radium-223 Response in Metastatic Castration-Resistant Prostate Cancer.
- Author
-
Saylor PJ, Otani K, Balza R, Ukleja J, Pleskow H, Fisher R, Kusaka E, Otani YS, Badusi PO, Smith MR, Meneely E, Olivier K, Lowe AC, Toner M, Maheswaran S, Haber DA, Yeap BY, Lee RJ, and Miyamoto DT
- Subjects
- Male, Humans, Receptors, Androgen, Prospective Studies, Biomarkers, Prostatic Neoplasms, Castration-Resistant diagnostic imaging, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant radiotherapy, Radium
- Abstract
Purpose: Radium-223 improves overall survival (OS) and reduces skeletal events in patients with bone metastatic castration-resistant prostate cancer (CRPC), but relevant biomarkers are lacking. We evaluated automated bone scan index (aBSI) and circulating tumor cell (CTC) analyses as potential biomarkers of prognosis and activity., Patients and Methods: Patients with bone metastatic CRPC were enrolled on a prospective single-arm study of standard radium-223.
99m Tc-MDP bone scan images at baseline, 2 months, and 6 months were quantitated using aBSI. CTCs at baseline, 1 month, and 2 months were enumerated and assessed for RNA expression of prostate cancer-specific genes using microfluidic enrichment followed by droplet digital polymerase chain reaction., Results: The median OS was 21.3 months in 22 patients. Lower baseline aBSI and minimal change in aBSI (<+0.7) from baseline to 2 months were each associated with better OS ( P = .00341 and P = .0139, respectively). The higher baseline CTC count of ≥5 CTC/7.5 mL was associated with worse OS (median, 10.1 v 32.9 months; P = .00568). CTCs declined at 2 months in four of 15 patients with detectable baseline CTCs. Among individual genes in CTCs, baseline expression of the splice variant AR-V7 was significantly associated with worse OS (hazard ratio, 5.20 [95% CI, 1.657 to 16.31]; P = .00195). Baseline detectable AR-V7 , higher aBSI, and CTC count ≥5 CTC/7.5 mL continued to have a significant independent negative impact on OS after controlling for prostate-specific antigen or alkaline phosphatase., Conclusion: Quantitative bone scan assessment with aBSI and CTC analyses are prognostic markers in patients treated with radium-223. AR-V7 expression in CTCs is a particularly promising prognostic biomarker and warrants validation in larger cohorts.- Published
- 2024
- Full Text
- View/download PDF
18. Deploying blood-based cancer screening.
- Author
-
Micalizzi DS, Sequist LV, and Haber DA
- Subjects
- Humans, Artificial Intelligence, Early Detection of Cancer methods, Mass Screening methods, Neoplasms blood, Neoplasms diagnosis, Blood Chemical Analysis methods
- Abstract
AI-based risk assessment may enable personalized blood-based multicancer screening.
- Published
- 2024
- Full Text
- View/download PDF
19. DrugMap: A quantitative pan-cancer analysis of cysteine ligandability.
- Author
-
Takahashi M, Chong HB, Zhang S, Lazarov MJ, Harry S, Maynard M, White R, Murrey HE, Hilbert B, Neil JR, Gohar M, Ge M, Zhang J, Durr BR, Kryukov G, Tsou CC, Brooijmans N, Alghali ASO, Rubio K, Vilanueva A, Harrison D, Koglin AS, Ojeda S, Karakyriakou B, Healy A, Assaad J, Makram F, Rachman I, Khandelwal N, Tien PC, Popoola G, Chen N, Vordermark K, Richter M, Patel H, Yang TY, Griesshaber H, Hosp T, van den Ouweland S, Hara T, Bussema L, Dong R, Shi L, Rasmussen MQ, Domingues AC, Lawless A, Fang J, Yoda S, Nguyen LP, Reeves SM, Wakefield FN, Acker A, Clark SE, Dubash T, Fisher DE, Maheswaran S, Haber DA, Boland G, Sade-Feldman M, Jenkins R, Hata A, Bardeesy N, Suva ML, Martin B, Liau B, Ott C, Rivera MN, Lawrence MS, and Bar-Peled L
- Abstract
Cysteine-focused chemical proteomic platforms have accelerated the clinical development of covalent inhibitors of a wide-range of targets in cancer. However, how different oncogenic contexts influence cysteine targeting remains unknown. To address this question, we have developed DrugMap , an atlas of cysteine ligandability compiled across 416 cancer cell lines. We unexpectedly find that cysteine ligandability varies across cancer cell lines, and we attribute this to differences in cellular redox states, protein conformational changes, and genetic mutations. Leveraging these findings, we identify actionable cysteines in NFκB1 and SOX10 and develop corresponding covalent ligands that block the activity of these transcription factors. We demonstrate that the NFκB1 probe blocks DNA binding, whereas the SOX10 ligand increases SOX10-SOX10 interactions and disrupts melanoma transcriptional signaling. Our findings reveal heterogeneity in cysteine ligandability across cancers, pinpoint cell-intrinsic features driving cysteine targeting, and illustrate the use of covalent probes to disrupt oncogenic transcription factor activity.
- Published
- 2023
- Full Text
- View/download PDF
20. Germline mutations and developmental mosaicism underlying EGFR -mutant lung cancer.
- Author
-
Burr R, Leshchiner I, Costantino CL, Blohmer M, Sundaresan T, Cha J, Seeger K, Guay S, Danysh BP, Gore I, Jacobs RA, Slowik K, Utro F, Rhrissorrakrai K, Levovitz C, Barth JL, Dubash T, Chirn B, Parida L, Sequist LV, Lennerz JK, Mino-Kenudson M, Maheswaran S, Naxerova K, Getz G, and Haber DA
- Abstract
While the development of multiple primary tumors in smokers with lung cancer can be attributed to carcinogen-induced field cancerization, the occurrence of multiple primary tumors in individuals with EGFR -mutant lung cancer who lack known environmental exposures remains unexplained. We identified ten patients with early-stage, resectable non-small cell lung cancer who presented with multiple anatomically distinct EGFR -mutant tumors. We analyzed the phylogenetic relationships among multiple tumors from each patient using whole exome sequencing (WES) and hypermutable poly-guanine (poly-G) repeat genotyping, as orthogonal methods for lineage tracing. In two patients, we identified germline EGFR variants, which confer moderately enhanced signaling when modeled in vitro . In four other patients, developmental mosaicism is supported by the poly-G lineage tracing and WES, indicating a common non-germline cell-of-origin. Thus, developmental mosaicism and germline variants define two distinct mechanisms of genetic predisposition to multiple EGFR -mutant primary tumors, with implications for understanding their etiology and clinical management.
- Published
- 2023
- Full Text
- View/download PDF
21. Modeling the novel SERD elacestrant in cultured fulvestrant-refractory HR-positive breast circulating tumor cells.
- Author
-
Dubash TD, Bardia A, Chirn B, Reeves BA, LiCausi JA, Burr R, Wittner BS, Rai S, Patel H, Bihani T, Arlt H, Bidard FC, Kaklamani VG, Aftimos P, Cortés J, Scartoni S, Fiascarelli A, Binaschi M, Habboubi N, Iafrate AJ, Toner M, Haber DA, and Maheswaran S
- Subjects
- Animals, Humans, Female, Fulvestrant, Receptors, Estrogen, Estrogen Antagonists therapeutic use, Disease Models, Animal, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Neoplastic Cells, Circulating
- Abstract
Purpose: Metastatic hormone receptor-positive (HR+) breast cancer initially responds to serial courses of endocrine therapy, but ultimately becomes refractory. Elacestrant, a new generation FDA-approved oral selective estrogen receptor degrader (SERD) and antagonist, has demonstrated efficacy in a subset of women with advanced HR+breast cancer, but there are few patient-derived models to characterize its effect in advanced cancers with diverse treatment histories and acquired mutations., Methods: We analyzed clinical outcomes with elacestrant, compared with endocrine therapy, among women who had previously been treated with a fulvestrant-containing regimen from the recent phase 3 EMERALD Study. We further modeled sensitivity to elacestrant, compared with the currently approved SERD, fulvestrant in patient-derived xenograft (PDX) models and cultured circulating tumor cells (CTCs)., Results: Analysis of the subset of breast cancer patients enrolled in the EMERALD study who had previously received a fulvestrant-containing regimen indicates that they had better progression-free survival with elacestrant than with standard-of-care endocrine therapy, a finding that was independent estrogen receptor (ESR1) gene mutations. We modeled elacestrant responsiveness using patient-derived xenograft (PDX) models and in ex vivo cultured CTCs derived from patients with HR+breast cancer extensively treated with multiple endocrine therapies, including fulvestrant. Both CTCs and PDX models are refractory to fulvestrant but sensitive to elacestrant, independent of mutations in ESR1 and Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha (PIK3CA) genes., Conclusion: Elacestrant retains efficacy in breast cancer cells that have acquired resistance to currently available ER targeting therapies. Elacestrant may be an option for patients with HR+/HER2- breast cancer whose disease progressed on fulvestrant in the metastatic setting., Translational Relevance: Serial endocrine therapy is the mainstay of management for metastatic HR+breast cancer, but acquisition of drug resistance highlights the need for better therapies. Elacestrant is a recently FDA-approved novel oral selective estrogen receptor degrader (SERD), with demonstrated efficacy in the EMERALD phase 3 clinical trial of refractory HR+breast cancer. Subgroup analysis of the EMERALD clinical trial identifies clinical benefit with elacestrant in patients who had received prior fulvestrant independent of the mutational status of the ESR1 gene, supporting its potential utility in treating refractory HR+breast cancer. Here, we use pre-clinical models, including ex vivo cultures of circulating tumor cells and patient-derived xenografts, to demonstrate the efficacy of elacestrant in breast cancer cells with acquired resistance to fulvestrant., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
22. A landscape of response to drug combinations in non-small cell lung cancer.
- Author
-
Nair NU, Greninger P, Zhang X, Friedman AA, Amzallag A, Cortez E, Sahu AD, Lee JS, Dastur A, Egan RK, Murchie E, Ceribelli M, Crowther GS, Beck E, McClanaghan J, Klump-Thomas C, Boisvert JL, Damon LJ, Wilson KM, Ho J, Tam A, McKnight C, Michael S, Itkin Z, Garnett MJ, Engelman JA, Haber DA, Thomas CJ, Ruppin E, and Benes CH
- Subjects
- Humans, Drug Combinations, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics
- Abstract
Combination of anti-cancer drugs is broadly seen as way to overcome the often-limited efficacy of single agents. The design and testing of combinations are however very challenging. Here we present a uniquely large dataset screening over 5000 targeted agent combinations across 81 non-small cell lung cancer cell lines. Our analysis reveals a profound heterogeneity of response across the tumor models. Notably, combinations very rarely result in a strong gain in efficacy over the range of response observable with single agents. Importantly, gain of activity over single agents is more often seen when co-targeting functionally proximal genes, offering a strategy for designing more efficient combinations. Because combinatorial effect is strongly context specific, tumor specificity should be achievable. The resource provided, together with an additional validation screen sheds light on major challenges and opportunities in building efficacious combinations against cancer and provides an opportunity for training computational models for synergy prediction., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. DNA hypomethylation silences anti-tumor immune genes in early prostate cancer and CTCs.
- Author
-
Guo H, Vuille JA, Wittner BS, Lachtara EM, Hou Y, Lin M, Zhao T, Raman AT, Russell HC, Reeves BA, Pleskow HM, Wu CL, Gnirke A, Meissner A, Efstathiou JA, Lee RJ, Toner M, Aryee MJ, Lawrence MS, Miyamoto DT, Maheswaran S, and Haber DA
- Subjects
- Animals, Humans, Male, Mice, Carcinogenesis genetics, DNA, Epigenesis, Genetic, Neoplastic Cells, Circulating, DNA Methylation, Prostatic Neoplasms genetics
- Abstract
Cancer is characterized by hypomethylation-associated silencing of large chromatin domains, whose contribution to tumorigenesis is uncertain. Through high-resolution genome-wide single-cell DNA methylation sequencing, we identify 40 core domains that are uniformly hypomethylated from the earliest detectable stages of prostate malignancy through metastatic circulating tumor cells (CTCs). Nested among these repressive domains are smaller loci with preserved methylation that escape silencing and are enriched for cell proliferation genes. Transcriptionally silenced genes within the core hypomethylated domains are enriched for immune-related genes; prominent among these is a single gene cluster harboring all five CD1 genes that present lipid antigens to NKT cells and four IFI16-related interferon-inducible genes implicated in innate immunity. The re-expression of CD1 or IFI16 murine orthologs in immuno-competent mice abrogates tumorigenesis, accompanied by the activation of anti-tumor immunity. Thus, early epigenetic changes may shape tumorigenesis, targeting co-located genes within defined chromosomal loci. Hypomethylation domains are detectable in blood specimens enriched for CTCs., Competing Interests: Declaration of interests Massachusetts General Hospital (MGH) has applied for patents regarding the CTC-iChip technology and CTC detection signatures. M.T., S.M., and D.A.H. are cofounders and have equity in Tell-Bio, which is not related to this work. The interests of these authors were reviewed and managed by MGH and Mass General Brigham (MGB) in accordance with their conflict of interest policies., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. A Gene Panel Associated With Abemaciclib Utility in ESR1 -Mutated Breast Cancer After Prior Cyclin-Dependent Kinase 4/6-Inhibitor Progression.
- Author
-
Brett JO, Dubash TD, Johnson GN, Niemierko A, Mariotti V, Kim LSL, Xi J, Pandey A, Dunne S, Nasrazadani A, Lloyd MR, Kambadakone A, Spring LM, Micalizzi DS, Onozato ML, Che D, Nayar U, Brufsky A, Kalinsky K, Ma CX, O'Shaughnessy J, Han HS, Iafrate AJ, Ryan LY, Juric D, Moy B, Ellisen LW, Maheswaran S, Wagle N, Haber DA, Bardia A, and Wander SA
- Subjects
- Humans, Female, Cyclin-Dependent Kinase 4 genetics, Retrospective Studies, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Disease Progression, Breast Neoplasms drug therapy, Breast Neoplasms genetics
- Abstract
Purpose: For patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC), first-line treatment is endocrine therapy (ET) plus cyclin-dependent kinase 4/6 inhibition (CDK4/6i). After disease progression, which often comes with ESR1 resistance mutations (ESR1-MUT), which therapies to use next and for which patients are open questions. An active area of exploration is treatment with further CDK4/6i, particularly abemaciclib, which has distinct pharmacokinetic and pharmacodynamic properties compared with the other approved CDK4/6 inhibitors, palbociclib and ribociclib. We investigated a gene panel to prognosticate abemaciclib susceptibility in patients with ESR1-MUT MBC after palbociclib progression., Methods: We examined a multicenter retrospective cohort of patients with ESR1-MUT MBC who received abemaciclib after disease progression on ET plus palbociclib. We generated a panel of CDK4/6i resistance genes and compared abemaciclib progression-free survival (PFS) in patients without versus with mutations in this panel (CDKi-R[-] v CDKi-R[+]). We studied how ESR1-MUT and CDKi-R mutations affect abemaciclib sensitivity of immortalized breast cancer cells and patient-derived circulating tumor cell lines in culture., Results: In ESR1-MUT MBC with disease progression on ET plus palbociclib, the median PFS was 7.0 months for CDKi-R(-) (n = 17) versus 3.5 months for CDKi-R(+) (n = 11), with a hazard ratio of 2.8 ( P = .03). In vitro, CDKi-R alterations but not ESR1-MUT induced abemaciclib resistance in immortalized breast cancer cells and were associated with resistance in circulating tumor cells., Conclusion: For ESR1-MUT MBC with resistance to ET and palbociclib, PFS on abemaciclib is longer for patients with CDKi-R(-) than CDKi-R(+). Although a small and retrospective data set, this is the first demonstration of a genomic panel associated with abemaciclib sensitivity in the postpalbociclib setting. Future directions include testing and improving this panel in additional data sets, to guide therapy selection for patients with HR+/HER2- MBC.
- Published
- 2023
- Full Text
- View/download PDF
25. TGF-β in the microenvironment induces a physiologically occurring immune-suppressive senescent state.
- Author
-
Matsuda S, Revandkar A, Dubash TD, Ravi A, Wittner BS, Lin M, Morris R, Burr R, Guo H, Seeger K, Szabolcs A, Che D, Nieman L, Getz GA, Ting DT, Lawrence MS, Gainor J, Haber DA, and Maheswaran S
- Subjects
- Mice, Animals, Humans, Phenotype, Disease Models, Animal, Cellular Microenvironment, Tumor Microenvironment, Cellular Senescence physiology, Transforming Growth Factor beta, Lung Neoplasms
- Abstract
TGF-β induces senescence in embryonic tissues. Whether TGF-β in the hypoxic tumor microenvironment (TME) induces senescence in cancer and how the ensuing senescence-associated secretory phenotype (SASP) remodels the cellular TME to influence immune checkpoint inhibitor (ICI) responses are unknown. We show that TGF-β induces a deeper senescent state under hypoxia than under normoxia; deep senescence correlates with the degree of E2F suppression and is marked by multinucleation, reduced reentry into proliferation, and a distinct 14-gene SASP. Suppressing TGF-β signaling in tumors in an immunocompetent mouse lung cancer model abrogates endogenous senescent cells and suppresses the 14-gene SASP and immune infiltration. Untreated human lung cancers with a high 14-gene SASP display immunosuppressive immune infiltration. In a lung cancer clinical trial of ICIs, elevated 14-gene SASP is associated with increased senescence, TGF-β and hypoxia signaling, and poor progression-free survival. Thus, TME-induced senescence may represent a naturally occurring state in cancer, contributing to an immune-suppressive phenotype associated with immune therapy resistance., Competing Interests: Declaration of interests A. Ravi has served as a consultant to Halo Solutions and Tyra Biosciences. J.F.G. has served as a compensated consultant or received honoraria from Bristol-Myers Squibb, Genentech/Roche, Takeda, Loxo/Lilly, Blueprint, Oncorus, Regeneron, Gilead, Moderna, Mirati, AstraZeneca, Pfizer, Novartis, iTeos, Nuvalent, Karyopharm, Beigene, Silverback Therapeutics, Merck, and GlydeBio; received research support from Novartis, Genentech/Roche, and Ariad/Takeda; received institutional research support from Bristol-Myers Squibb, Tesaro, Moderna, Blueprint, Jounce, Array Biopharma, Merck, Adaptimmune, Novartis, and Alexo; and has an immediate family member who is an employee with equity at Ironwood Pharmaceuticals. D.T.T. has received consulting fees from ROME Therapeutics, Tekla Capital, Ikena Oncology, Foundation Medicine, Inc., NanoString Technologies, and Pfizer that are not related to this work. D.T.T. is a founder of and has equity in ROME Therapeutics, PanTher Therapeutics, and TellBio, Inc., which are not related to this work. D.T.T. receives research support from ACD-Biotechne, PureTech Health LLC, and Ribon Therapeutics, which was not used in this work. D.T.T.’s interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict-of-interest policies. S. Maheswaran and D.A.H. are co-founders and have equity in Tell-Bio, which is not related to this work., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Downregulation of KEAP1 in melanoma promotes resistance to immune checkpoint blockade.
- Author
-
Fox DB, Ebright RY, Hong X, Russell HC, Guo H, LaSalle TJ, Wittner BS, Poux N, Vuille JA, Toner M, Hacohen N, Boland GM, Sen DR, Sullivan RJ, Maheswaran S, and Haber DA
- Abstract
Immune checkpoint blockade (ICB) has demonstrated efficacy in patients with melanoma, but many exhibit poor responses. Using single cell RNA sequencing of melanoma patient-derived circulating tumor cells (CTCs) and functional characterization using mouse melanoma models, we show that the KEAP1/NRF2 pathway modulates sensitivity to ICB, independently of tumorigenesis. The NRF2 negative regulator, KEAP1, shows intrinsic variation in expression, leading to tumor heterogeneity and subclonal resistance., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
27. Targeting breast and pancreatic cancer metastasis using a dual-cadherin antibody.
- Author
-
Micalizzi DS, Che D, Nicholson BT, Edd JF, Desai N, Lang ER, Toner M, Maheswaran S, Ting DT, and Haber DA
- Subjects
- Humans, Animals, Mice, Female, Epithelial-Mesenchymal Transition, Cell Line, Tumor, Cadherins metabolism, Neoplastic Processes, Mice, Nude, Mice, SCID, Epitopes, Neoplasm Metastasis, Neoplastic Cells, Circulating pathology, Pancreatic Neoplasms drug therapy, Breast Neoplasms drug therapy
- Abstract
The successful application of antibody-based therapeutics in either primary or metastatic cancer depends upon the selection of rare cell surface epitopes that distinguish cancer cells from surrounding normal epithelial cells. By contrast, as circulating tumor cells (CTCs) transit through the bloodstream, they are surrounded by hematopoietic cells with dramatically distinct cell surface proteins, greatly expanding the number of targetable epitopes. Here, we show that an antibody (23C6) against cadherin proteins effectively suppresses blood-borne metastasis in mouse isogenic and xenograft models of triple negative breast and pancreatic cancers. The 23C6 antibody is remarkable in that it recognizes both the epithelial E-cadherin (CDH1) and mesenchymal OB-cadherin (CDH11), thus overcoming considerable heterogeneity across tumor cells. Despite its efficacy against single cells in circulation, the antibody does not suppress primary tumor formation, nor does it elicit detectable toxicity in normal epithelial organs, where cadherins may be engaged within intercellular junctions and hence inaccessible for antibody binding. Antibody-mediated suppression of metastasis is comparable in matched immunocompetent and immunodeficient mouse models. Together, these studies raise the possibility of antibody targeting CTCs within the vasculature, thereby suppressing blood-borne metastasis.
- Published
- 2022
- Full Text
- View/download PDF
28. Isolation of circulating tumor cells.
- Author
-
Edd JF, Mishra A, Smith KC, Kapur R, Maheswaran S, Haber DA, and Toner M
- Abstract
Circulating tumor cells (CTCs) enter the vasculature from solid tumors and disseminate widely to initiate metastases. Mining the metastatic-enriched molecular signatures of CTCs before, during, and after treatment holds unique potential in personalized oncology. Their extreme rarity, however, requires isolation from large blood volumes at high yield and purity, yet they overlap leukocytes in size and other biophysical properties. Additionally, many CTCs lack EpCAM that underlies much of affinity-based capture, complicating their separation from blood. Here, we provide a comprehensive introduction of CTC isolation technology, by analyzing key separation modes and integrated isolation strategies. Attention is focused on recent progress in microfluidics, where an accelerating evolution is occurring in high-throughput sorting of cells along multiple dimensions., Competing Interests: M.G.H. filed for patent protection for the CTC-iChip technology. M.T., R.K., D.H., and S.M. are founders of TellBio who are commercializing using the CTC-iChip 2. The authors declare that they have no other competing interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
29. Differential Kinase Activity Across Prostate Tumor Compartments Defines Sensitivity to Target Inhibition.
- Author
-
Karabacak NM, Zheng Y, Dubash TD, Burr R, Micalizzi DS, Wittner BS, Lin M, Wiley DF, Comaills V, Emmons E, Niederhoffer KL, Ho U, Ukleja J, Che D, Stowe H, Nieman LT, Haas W, Stott SL, Lawrence MS, Ting DT, Miyamoto DT, Haber DA, Toner M, and Maheswaran S
- Subjects
- Animals, Cell Line, Tumor, Epithelial Cell Adhesion Molecule, Humans, Male, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors pharmacology, TOR Serine-Threonine Kinases metabolism, Tumor Microenvironment, Prostatic Neoplasms drug therapy, Proteomics
- Abstract
Cancer therapy often results in heterogeneous responses in different metastatic lesions in the same patient. Inter- and intratumor heterogeneity in signaling within various tumor compartments and its impact on therapy are not well characterized due to the limited sensitivity of single-cell proteomic approaches. To overcome this barrier, we applied single-cell mass cytometry with a customized 26-antibody panel to PTEN-deleted orthotopic prostate cancer xenograft models to measure the evolution of kinase activities in different tumor compartments during metastasis or drug treatment. Compared with primary tumors and circulating tumor cells (CTC), bone metastases, but not lung and liver metastases, exhibited elevated PI3K/mTOR signaling and overexpressed receptor tyrosine kinases (RTK) including c-MET protein. Suppression of c-MET impaired tumor growth in the bone. Intratumoral heterogeneity within tumor compartments also arose from highly proliferative EpCAM-high epithelial cells with increased PI3K and mTOR kinase activities coexisting with poorly proliferating EpCAM-low mesenchymal populations with reduced kinase activities; these findings were recapitulated in epithelial and mesenchymal CTC populations in patients with metastatic prostate and breast cancer. Increased kinase activity in EpCAM-high cells rendered them more sensitive to PI3K/mTOR inhibition, and drug-resistant EpCAM-low populations with reduced kinase activity emerged over time. Taken together, single-cell proteomics indicate that microenvironment- and cell state-dependent activation of kinase networks create heterogeneity and differential drug sensitivity among and within tumor populations across different sites, defining a new paradigm of drug responses to kinase inhibitors., Significance: Single-cell mass cytometry analyses provide insights into the differences in kinase activities across tumor compartments and cell states, which contribute to heterogeneous responses to targeted therapies., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
30. Negative-Selection Enrichment of Circulating Tumor Cells from Peripheral Blood Using the Microfluidic CTC-iChip.
- Author
-
Burr R, Edd JF, Chirn B, Mishra A, Haber DA, Toner M, and Maheswaran S
- Subjects
- Cell Count, Cell Line, Tumor, Cell Separation methods, Humans, Microfluidics methods, Neoplastic Cells, Circulating pathology
- Abstract
The ability to isolate and analyze rare circulating tumor cells (CTCs) holds the potential to increase our understanding of cancer evolution and allows monitoring of disease and therapeutic responses through a relatively non-invasive blood-based biopsy. While many methods have been described to isolate CTCs from the blood, the vast majority rely on size-based sorting or positive selection of CTCs based on surface markers, which introduces bias into the downstream product by making assumptions about these heterogenous cells. Here we describe a negative-selection protocol for enrichment of CTCs through removal of blood components including red blood cells, platelets, and white blood cells. This procedure results in a product that is amenable to downstream single-cell analytics including RNA-Seq, ATAC-Seq and DNA methylation, droplet digital PCR (ddPCR) for tumor specific transcripts, staining and extensive image analysis, and ex vivo culture of patient-derived CTCs., (© 2022. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
31. NR4A1 regulates expression of immediate early genes, suppressing replication stress in cancer.
- Author
-
Guo H, Golczer G, Wittner BS, Langenbucher A, Zachariah M, Dubash TD, Hong X, Comaills V, Burr R, Ebright RY, Horwitz E, Vuille JA, Hajizadeh S, Wiley DF, Reeves BA, Zhang JM, Niederhoffer KL, Lu C, Wesley B, Ho U, Nieman LT, Toner M, Vasudevan S, Zou L, Mostoslavsky R, Maheswaran S, Lawrence MS, and Haber DA
- Subjects
- 3' Untranslated Regions, Animals, Antineoplastic Agents pharmacology, Binding Sites, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Chromatin Assembly and Disassembly, Female, Gene Expression Regulation, Neoplastic, Genomic Instability, HEK293 Cells, Humans, Immediate-Early Proteins genetics, Indoles pharmacology, MCF-7 Cells, Mice, Inbred NOD, Mice, SCID, Neoplastic Cells, Circulating drug effects, Neoplastic Cells, Circulating pathology, Nuclear Receptor Subfamily 4, Group A, Member 1 antagonists & inhibitors, Nuclear Receptor Subfamily 4, Group A, Member 1 genetics, Phenylacetates pharmacology, Proto-Oncogene Proteins c-fos genetics, Proto-Oncogene Proteins c-fos metabolism, R-Loop Structures, RNA Polymerase II genetics, RNA Polymerase II metabolism, Signal Transduction, Transcription Elongation, Genetic, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Mice, Breast Neoplasms metabolism, Cell Proliferation drug effects, Immediate-Early Proteins metabolism, Mitosis drug effects, Neoplastic Cells, Circulating metabolism, Nuclear Receptor Subfamily 4, Group A, Member 1 metabolism
- Abstract
Deregulation of oncogenic signals in cancer triggers replication stress. Immediate early genes (IEGs) are rapidly and transiently expressed following stressful signals, contributing to an integrated response. Here, we find that the orphan nuclear receptor NR4A1 localizes across the gene body and 3' UTR of IEGs, where it inhibits transcriptional elongation by RNA Pol II, generating R-loops and accessible chromatin domains. Acute replication stress causes immediate dissociation of NR4A1 and a burst of transcriptionally poised IEG expression. Ectopic expression of NR4A1 enhances tumorigenesis by breast cancer cells, while its deletion leads to massive chromosomal instability and proliferative failure, driven by deregulated expression of its IEG target, FOS. Approximately half of breast and other primary cancers exhibit accessible chromatin domains at IEG gene bodies, consistent with this stress-regulatory pathway. Cancers that have retained this mechanism in adapting to oncogenic replication stress may be dependent on NR4A1 for their proliferation., Competing Interests: Declaration of interests Massachusetts General Hospital (MGH) has applied for patents regarding the CTC-iChip technology and CTC detection signatures. M.T., D.A.H., and S.M. are cofounders and have equity in Tell-Bio, which is not related to this work. The interests of these authors were reviewed and managed by MGH and Partners HealthCare in accordance with their conflict of interest policies. All other authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Evaluation of endocrine resistance using ESR1 genotyping of circulating tumor cells and plasma DNA.
- Author
-
Sundaresan TK, Dubash TD, Zheng Z, Bardia A, Wittner BS, Aceto N, Silva EJ, Fox DB, Liebers M, Kapur R, Iafrate J, Toner M, Maheswaran S, and Haber DA
- Subjects
- Estrogen Receptor alpha genetics, Female, Genotype, Humans, Mutation, Neoplasm Recurrence, Local, Prospective Studies, Breast Neoplasms, Circulating Tumor DNA, Neoplastic Cells, Circulating
- Abstract
Purpose: Therapeutic efficacy of hormonal therapies to target estrogen receptor (ER)-positive breast cancer is limited by the acquisition of ligand-independent ESR1 mutations, which confer treatment resistance to aromatase inhibitors (AIs). Monitoring for the emergence of such mutations may enable individualized therapy. We thus assessed CTC- and ctDNA-based detection of ESR1 mutations with the aim of evaluating non-invasive approaches for the determination of endocrine resistance., Patients and Methods: In a prospective cohort of 55 women with hormone receptor-positive metastatic breast cancer, we isolated circulating tumor cells (CTCs) and developed a high-sensitivity method for the detection of ESR1 mutations in these CTCs. In patients with sufficient plasma for the simultaneous extraction of circulating tumor DNA (ctDNA), we performed a parallel analysis of ESR1 mutations using multiplex droplet digital PCR (ddPCR) and examined the agreement between these two platforms. Finally, we isolated single CTCs from a subset of these patients and reviewed RNA expression to explore alternate methods of evaluating endocrine responsiveness., Results: High-sensitivity ESR1 sequencing from CTCs revealed mono- and oligoclonal mutations in 22% of patients. These were concordant with plasma DNA sequencing in 95% of cases. Emergence of ESR1 mutations was correlated both with time to metastatic relapse and duration of AI therapy following such recurrence. The Presence of an ESR1 mutation, compared to ESR1 wild type, was associated with markedly shorter Progression-Free Survival on AI-based therapies (p = 0.0006), but unaltered to other non-AI-based therapies (p = 0.73). Compared with ESR1 mutant cases, AI-resistant CTCs with wild-type ESR1 showed an elevated ER-coactivator RNA signature, consistent with their predicted response to second-line hormonal therapies., Conclusion: Blood-based serial monitoring may guide the selection of precision therapeutics for women with AI-resistant ER-positive breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
33. RNA transcripts stimulate homologous recombination by forming DR-loops.
- Author
-
Ouyang J, Yadav T, Zhang JM, Yang H, Rheinbay E, Guo H, Haber DA, Lan L, and Zou L
- Subjects
- Basic Helix-Loop-Helix Transcription Factors genetics, Cell Line, DNA chemistry, DNA Breaks, Double-Stranded, DNA Repair, DNA-Binding Proteins metabolism, Genes genetics, Genes, Reporter genetics, Green Fluorescent Proteins genetics, Humans, In Vitro Techniques, RNA, Messenger chemistry, RNA-Binding Proteins metabolism, Rad51 Recombinase metabolism, DNA genetics, DNA metabolism, Homologous Recombination genetics, R-Loop Structures genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Transcription, Genetic
- Abstract
Homologous recombination (HR) repairs DNA double-strand breaks (DSBs) in the S and G2 phases of the cell cycle
1-3 . Several HR proteins are preferentially recruited to DSBs at transcriptionally active loci4-10 , but how transcription promotes HR is poorly understood. Here we develop an assay to assess the effect of local transcription on HR. Using this assay, we find that transcription stimulates HR to a substantial extent. Tethering RNA transcripts to the vicinity of DSBs recapitulates the effects of local transcription, which suggests that transcription enhances HR through RNA transcripts. Tethered RNA transcripts stimulate HR in a sequence- and orientation-dependent manner, indicating that they function by forming DNA-RNA hybrids. In contrast to most HR proteins, RAD51-associated protein 1 (RAD51AP1) only promotes HR when local transcription is active. RAD51AP1 drives the formation of R-loops in vitro and is required for tethered RNAs to stimulate HR in cells. Notably, RAD51AP1 is necessary for the DSB-induced formation of DNA-RNA hybrids in donor DNA, linking R-loops to D-loops. In vitro, RAD51AP1-generated R-loops enhance the RAD51-mediated formation of D-loops locally and give rise to intermediates that we term 'DR-loops', which contain both DNA-DNA and DNA-RNA hybrids and favour RAD51 function. Thus, at DSBs in transcribed regions, RAD51AP1 promotes the invasion of RNA transcripts into donor DNA, and stimulates HR through the formation of DR-loops.- Published
- 2021
- Full Text
- View/download PDF
34. The Lipogenic Regulator SREBP2 Induces Transferrin in Circulating Melanoma Cells and Suppresses Ferroptosis.
- Author
-
Hong X, Roh W, Sullivan RJ, Wong KHK, Wittner BS, Guo H, Dubash TD, Sade-Feldman M, Wesley B, Horwitz E, Boland GM, Marvin DL, Bonesteel T, Lu C, Aguet F, Burr R, Freeman SS, Parida L, Calhoun K, Jewett MK, Nieman LT, Hacohen N, Näär AM, Ting DT, Toner M, Stott SL, Getz G, Maheswaran S, and Haber DA
- Subjects
- Biomarkers, Tumor, Cells, Cultured, Disease Susceptibility, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Melanoma pathology, Mutation, Neoplastic Cells, Circulating pathology, Signal Transduction, Single-Cell Analysis, Sterol Regulatory Element Binding Protein 2 metabolism, Ferroptosis genetics, Lipogenesis genetics, Melanoma genetics, Melanoma metabolism, Neoplastic Cells, Circulating metabolism, Sterol Regulatory Element Binding Protein 2 genetics, Transferrin metabolism
- Abstract
Circulating tumor cells (CTC) are shed by cancer into the bloodstream, where a viable subset overcomes oxidative stress to initiate metastasis. We show that single CTCs from patients with melanoma coordinately upregulate lipogenesis and iron homeostasis pathways. These are correlated with both intrinsic and acquired resistance to BRAF inhibitors across clonal cultures of BRAF -mutant CTCs. The lipogenesis regulator SREBP2 directly induces transcription of the iron carrier Transferrin ( TF ), reducing intracellular iron pools, reactive oxygen species, and lipid peroxidation, thereby conferring resistance to inducers of ferroptosis. Knockdown of endogenous TF impairs tumor formation by melanoma CTCs, and their tumorigenic defects are partially rescued by the lipophilic antioxidants ferrostatin-1 and vitamin E. In a prospective melanoma cohort, presence of CTCs with high lipogenic and iron metabolic RNA signatures is correlated with adverse clinical outcome, irrespective of treatment regimen. Thus, SREBP2-driven iron homeostatic pathways contribute to cancer progression, drug resistance, and metastasis. SIGNIFICANCE: Through single-cell analysis of primary and cultured melanoma CTCs, we have uncovered intrinsic cancer cell heterogeneity within lipogenic and iron homeostatic pathways that modulates resistance to BRAF inhibitors and to ferroptosis inducers. Activation of these pathways within CTCs is correlated with adverse clinical outcome, pointing to therapeutic opportunities. This article is highlighted in the In This Issue feature, p. 521 ., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
35. Translational Regulation of Cancer Metastasis.
- Author
-
Micalizzi DS, Ebright RY, Haber DA, and Maheswaran S
- Subjects
- Carcinogenesis metabolism, Cell Movement, Cell Survival physiology, Humans, Neoplasm Metastasis pathology, Neoplasm Metastasis therapy, Neoplasm Proteins biosynthesis, Neoplasms therapy, Neovascularization, Pathologic etiology, Phenotype, TOR Serine-Threonine Kinases metabolism, Transforming Growth Factor beta metabolism, Tumor Escape physiology, Tumor Hypoxia physiology, Tumor Microenvironment physiology, Epithelial-Mesenchymal Transition physiology, Neoplasm Metastasis genetics, Protein Biosynthesis physiology
- Abstract
Deregulation of the mRNA translational process has been observed during tumorigenesis. However, recent findings have shown that deregulation of translation also contributes specifically to cancer cell spread. During metastasis, cancer cells undergo changes in cellular state, permitting the acquisition of features necessary for cell survival, dissemination, and outgrowth. In addition, metastatic cells respond to external cues, allowing for their persistence under significant cellular and microenvironmental stresses. Recent work has revealed the importance of mRNA translation to these dynamic changes, including regulation of cell states through epithelial-to-mesenchymal transition and tumor dormancy and as a response to external stresses such as hypoxia and immune surveillance. In this review, we focus on examples of altered translation underlying these phenotypic changes and responses to external cues and explore how they contribute to metastatic progression. We also highlight the therapeutic opportunities presented by aberrant mRNA translation, suggesting novel ways to target metastatic tumor cells., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
36. HIF1A signaling selectively supports proliferation of breast cancer in the brain.
- Author
-
Ebright RY, Zachariah MA, Micalizzi DS, Wittner BS, Niederhoffer KL, Nieman LT, Chirn B, Wiley DF, Wesley B, Shaw B, Nieblas-Bedolla E, Atlas L, Szabolcs A, Iafrate AJ, Toner M, Ting DT, Brastianos PK, Haber DA, and Maheswaran S
- Subjects
- Animals, Brain Neoplasms blood, Brain Neoplasms mortality, Breast Neoplasms blood, Breast Neoplasms mortality, Cell Hypoxia, Cell Proliferation, Female, Gene Expression Regulation, Neoplastic, Humans, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Mammary Glands, Animal pathology, Metabolomics, Mice, RNA, Small Interfering metabolism, RNA-Seq, Signal Transduction drug effects, Signal Transduction genetics, Spheroids, Cellular, Stereotaxic Techniques, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Brain pathology, Brain Neoplasms secondary, Breast Neoplasms pathology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neoplastic Cells, Circulating metabolism
- Abstract
Blood-borne metastasis to the brain is a major complication of breast cancer, but cellular pathways that enable cancer cells to selectively grow in the brain microenvironment are poorly understood. We find that cultured circulating tumor cells (CTCs), derived from blood samples of women with advanced breast cancer and directly inoculated into the mouse frontal lobe, exhibit striking differences in proliferative potential in the brain. Derivative cell lines generated by serial intracranial injections acquire selectively increased proliferative competency in the brain, with reduced orthotopic tumor growth. Increased Hypoxia Inducible Factor 1A (HIF1A)-associated signaling correlates with enhanced proliferation in the brain, and shRNA-mediated suppression of HIF1A or drug inhibition of HIF-associated glycolytic pathways selectively impairs brain tumor growth while minimally impacting mammary tumor growth. In clinical specimens, brain metastases have elevated HIF1A protein expression, compared with matched primary breast tumors, and in patients with brain metastases, hypoxic signaling within CTCs predicts decreased overall survival. The selective activation of hypoxic signaling by metastatic breast cancer in the brain may have therapeutic implications.
- Published
- 2020
- Full Text
- View/download PDF
37. Identification of Somatically Acquired BRCA1/2 Mutations by cfDNA Analysis in Patients with Metastatic Breast Cancer.
- Author
-
Vidula N, Dubash T, Lawrence MS, Simoneau A, Niemierko A, Blouch E, Nagy B, Roh W, Chirn B, Reeves BA, Malvarosa G, Lennerz J, Isakoff SJ, Juric D, Micalizzi D, Wander S, Spring L, Moy B, Shannon K, Younger J, Lanman R, Toner M, Iafrate AJ, Getz G, Zou L, Ellisen LW, Maheswaran S, Haber DA, and Bardia A
- Subjects
- Aged, Breast Neoplasms blood, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Line, Tumor, Circulating Tumor DNA blood, DNA Mutational Analysis, Drug Resistance, Neoplasm genetics, Female, High-Throughput Nucleotide Sequencing, Humans, Middle Aged, Mutation, Neoplastic Cells, Circulating pathology, Phthalazines pharmacology, Phthalazines therapeutic use, Piperazines pharmacology, Piperazines therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Retrospective Studies, Exome Sequencing, BRCA1 Protein genetics, BRCA2 Protein genetics, Breast Neoplasms genetics, Circulating Tumor DNA genetics
- Abstract
Purpose: Plasma genotyping may identify mutations in potentially "actionable" cancer genes, such as BRCA1/2 , but their clinical significance is not well-defined. We evaluated the characteristics of somatically acquired BRCA1/2 mutations in patients with metastatic breast cancer (MBC)., Experimental Design: Patients with MBC undergoing routine cell-free DNA (cfDNA) next-generation sequencing (73-gene panel) before starting a new therapy were included. Somatic BRCA1/2 mutations were classified as known germline pathogenic mutations or novel variants, and linked to clinicopathologic characteristics. The effect of the PARP inhibitor, olaparib, was assessed in vitro , using cultured circulating tumor cells (CTCs) from a patient with a somatically acquired BRCA1 mutation and a second patient with an acquired BRCA2 mutation., Results: Among 215 patients with MBC, 29 (13.5%) had somatic cfDNA BRCA1/2 mutations [nine (4%) known germline pathogenic and rest (9%) novel variants]. Known germline pathogenic BRCA1/2 mutations were common in younger patients ( P = 0.008), those with triple-negative disease ( P = 0.022), and they were more likely to be protein-truncating alterations and be associated with TP53 mutations. Functional analysis of a CTC culture harboring a somatic BRCA1 mutation demonstrated high sensitivity to PARP inhibition, while another CTC culture harboring a somatic BRCA2 mutation showed no differential sensitivity. Across the entire cohort, APOBEC mutational signatures (COSMIC Signatures 2 and 13) and the "BRCA" mutational signature (COSMIC Signature 3) were present in BRCA1/2- mutant and wild-type cases, demonstrating the high mutational burden associated with advanced MBC., Conclusions: Somatic BRCA1/2 mutations are readily detectable in MBC by cfDNA analysis, and may be present as both known germline pathogenic and novel variants., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
38. Ultrahigh-throughput magnetic sorting of large blood volumes for epitope-agnostic isolation of circulating tumor cells.
- Author
-
Mishra A, Dubash TD, Edd JF, Jewett MK, Garre SG, Karabacak NM, Rabe DC, Mutlu BR, Walsh JR, Kapur R, Stott SL, Maheswaran S, Haber DA, and Toner M
- Subjects
- Cell Line, Tumor, Cell Separation instrumentation, High-Throughput Screening Assays instrumentation, Humans, Leukapheresis methods, Magnetic Fields, Microfluidics instrumentation, Cell Separation methods, High-Throughput Screening Assays methods, Microfluidics methods, Neoplastic Cells, Circulating classification
- Abstract
Circulating tumor cell (CTC)-based liquid biopsies provide unique opportunities for cancer diagnostics, treatment selection, and response monitoring, but even with advanced microfluidic technologies for rare cell detection the very low number of CTCs in standard 10-mL peripheral blood samples limits their clinical utility. Clinical leukapheresis can concentrate mononuclear cells from almost the entire blood volume, but such large numbers and concentrations of cells are incompatible with current rare cell enrichment technologies. Here, we describe an ultrahigh-throughput microfluidic chip,
LP CTC-iChip, that rapidly sorts through an entire leukapheresis product of over 6 billion nucleated cells, increasing CTC isolation capacity by two orders of magnitude (86% recovery with 105 enrichment). Using soft iron-filled channels to act as magnetic microlenses, we intensify the field gradient within sorting channels. Increasing magnetic fields applied to inertially focused streams of cells effectively deplete massive numbers of magnetically labeled leukocytes within microfluidic channels. The negative depletion of antibody-tagged leukocytes enables isolation of potentially viable CTCs without bias for expression of specific tumor epitopes, making this platform applicable to all solid tumors. Thus, the initial enrichment by routine leukapheresis of mononuclear cells from very large blood volumes, followed by rapid flow, high-gradient magnetic sorting of untagged CTCs, provides a technology for noninvasive isolation of cancer cells in sufficient numbers for multiple clinical and experimental applications., Competing Interests: Competing interest statement: Massachusetts General Hospital has filed patent applications for the circulating tumor cell isolation technology described in this publication., (Copyright © 2020 the Author(s). Published by PNAS.)- Published
- 2020
- Full Text
- View/download PDF
39. Pancreatic circulating tumor cell profiling identifies LIN28B as a metastasis driver and drug target.
- Author
-
Franses JW, Philipp J, Missios P, Bhan I, Liu A, Yashaswini C, Tai E, Zhu H, Ligorio M, Nicholson B, Tassoni EM, Desai N, Kulkarni AS, Szabolcs A, Hong TS, Liss AS, Fernandez-Del Castillo C, Ryan DP, Maheswaran S, Haber DA, Daley GQ, and Ting DT
- Subjects
- Adult, Aged, Animals, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal metabolism, Cell Line, Tumor, Cell Movement genetics, Female, Gene Expression Regulation, Neoplastic, HMGA2 Protein metabolism, Humans, Kaplan-Meier Estimate, Kruppel-Like Factor 4, Male, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Middle Aged, Neoplasm Metastasis, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, RNA-Binding Proteins metabolism, Carcinoma, Pancreatic Ductal genetics, HMGA2 Protein genetics, MicroRNAs genetics, Neoplastic Cells, Circulating metabolism, Pancreatic Neoplasms genetics, RNA-Binding Proteins genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) lethality is due to metastatic dissemination. Characterization of rare, heterogeneous circulating tumor cells (CTCs) can provide insight into metastasis and guide development of novel therapies. Using the CTC-iChip to purify CTCs from PDAC patients for RNA-seq characterization, we identify three major correlated gene sets, with stemness genes LIN28B/KLF4, WNT5A, and LGALS3 enriched in each correlated gene set; only LIN28B CTC expression was prognostic. CRISPR knockout of LIN28B-an oncofetal RNA-binding protein exerting diverse effects via negative regulation of let-7 miRNAs and other RNA targets-in cell and animal models confers a less aggressive/metastatic phenotype. This correlates with de-repression of let-7 miRNAs and is mimicked by silencing of downstream let-7 target HMGA2 or chemical inhibition of LIN28B/let-7 binding. Molecular characterization of CTCs provides a unique opportunity to correlated gene set metastatic profiles, identify drivers of dissemination, and develop therapies targeting the "seeds" of metastasis.
- Published
- 2020
- Full Text
- View/download PDF
40. ATRAID regulates the action of nitrogen-containing bisphosphonates on bone.
- Author
-
Surface LE, Burrow DT, Li J, Park J, Kumar S, Lyu C, Song N, Yu Z, Rajagopal A, Bae Y, Lee BH, Mumm S, Gu CC, Baker JC, Mohseni M, Sum M, Huskey M, Duan S, Bijanki VN, Civitelli R, Gardner MJ, McAndrew CM, Ricci WM, Gurnett CA, Diemer K, Wan F, Costantino CL, Shannon KM, Raje N, Dodson TB, Haber DA, Carette JE, Varadarajan M, Brummelkamp TR, Birsoy K, Sabatini DM, Haller G, and Peterson TR
- Subjects
- Alendronate pharmacology, Animals, Bone and Bones, Humans, Mice, Osteoclasts, Diphosphonates pharmacology, Diphosphonates therapeutic use, Nitrogen
- Abstract
Nitrogen-containing bisphosphonates (N-BPs), such as alendronate, are the most widely prescribed medications for diseases involving bone, with nearly 200 million prescriptions written annually. Recently, widespread use of N-BPs has been challenged due to the risk of rare but traumatic side effects such as atypical femoral fracture (AFF) and osteonecrosis of the jaw (ONJ). N-BPs bind to and inhibit farnesyl diphosphate synthase, resulting in defects in protein prenylation. Yet, it remains poorly understood what other cellular factors might allow N-BPs to exert their pharmacological effects. Here, we performed genome-wide studies in cells and patients to identify the poorly characterized gene, ATRAID Loss of ATRAID function results in selective resistance to N-BP-mediated loss of cell viability and the prevention of alendronate-mediated inhibition of prenylation. ATRAID is required for alendronate inhibition of osteoclast function, and ATRAID -deficient mice have impaired therapeutic responses to alendronate in both postmenopausal and senile (old age) osteoporosis models. Last, we performed exome sequencing on patients taking N-BPs that suffered ONJ or an AFF. ATRAID is one of three genes that contain rare nonsynonymous coding variants in patients with ONJ or an AFF that is also differentially expressed in poor outcome groups of patients treated with N-BPs. We functionally validated this patient variation in ATRAID as conferring cellular hypersensitivity to N-BPs. Our work adds key insight into the mechanistic action of N-BPs and the processes that might underlie differential responsiveness to N-BPs in people., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
41. In-flow measurement of cell-cell adhesion using oscillatory inertial microfluidics.
- Author
-
Mutlu BR, Dubash T, Dietsche C, Mishra A, Ozbey A, Keim K, Edd JF, Haber DA, Maheswaran S, and Toner M
- Subjects
- Cell Adhesion, Humans, Lab-On-A-Chip Devices, Neoplastic Cells, Circulating pathology, Oscillometry
- Abstract
Multicellular clusters in circulation can exhibit a substantially different function and biomarker significance compared to individual cells. Notably, clusters of circulating tumor cells (CTCs) are much more effective initiators of metastasis than single CTCs, and correlate with worse patient prognoses. Measuring the cell-cell adhesion strength of CTC clusters is a critical step towards understanding their subsistence in the circulation and mechanism of elevated tumorigenicity. However, measuring cell-cell adhesion forces in flow is elusive using existing methods. Here, we report an oscillatory inertial microfluidics system which exerts a repeating fluidic force profile on suspended cell doublets to determine their cell-cell adhesion strength (F
s ), without any biophysical modifications to the cell surface and physiological morphology. Using our system, we analyzed a large number (N > 500) of doublets from a patient-derived breast cancer CTC line. We discovered that the cell-cell adhesion strength of CTC doublets varied almost 20-fold between the weakly adhered (Fs < 28 nN) and strongly bound subpopulations (Fs > 542 nN). Our system can be used with other cancer or noncancer cells without restrictions, and may be used for rapid screening of drugs aiming to disrupt the highly-metastatic CTC clusters in circulation.- Published
- 2020
- Full Text
- View/download PDF
42. Integrating genomic features for non-invasive early lung cancer detection.
- Author
-
Chabon JJ, Hamilton EG, Kurtz DM, Esfahani MS, Moding EJ, Stehr H, Schroers-Martin J, Nabet BY, Chen B, Chaudhuri AA, Liu CL, Hui AB, Jin MC, Azad TD, Almanza D, Jeon YJ, Nesselbush MC, Co Ting Keh L, Bonilla RF, Yoo CH, Ko RB, Chen EL, Merriott DJ, Massion PP, Mansfield AS, Jen J, Ren HZ, Lin SH, Costantino CL, Burr R, Tibshirani R, Gambhir SS, Berry GJ, Jensen KC, West RB, Neal JW, Wakelee HA, Loo BW Jr, Kunder CA, Leung AN, Lui NS, Berry MF, Shrager JB, Nair VS, Haber DA, Sequist LV, Alizadeh AA, and Diehn M
- Subjects
- Cohort Studies, Female, Hematopoiesis genetics, Humans, Lung metabolism, Lung pathology, Lung Neoplasms blood, Lung Neoplasms pathology, Male, Middle Aged, Reproducibility of Results, Circulating Tumor DNA analysis, Circulating Tumor DNA genetics, Early Detection of Cancer methods, Genome, Human genetics, Lung Neoplasms diagnosis, Lung Neoplasms genetics, Mutation
- Abstract
Radiologic screening of high-risk adults reduces lung-cancer-related mortality
1,2 ; however, a small minority of eligible individuals undergo such screening in the United States3,4 . The availability of blood-based tests could increase screening uptake. Here we introduce improvements to cancer personalized profiling by deep sequencing (CAPP-Seq)5 , a method for the analysis of circulating tumour DNA (ctDNA), to better facilitate screening applications. We show that, although levels are very low in early-stage lung cancers, ctDNA is present prior to treatment in most patients and its presence is strongly prognostic. We also find that the majority of somatic mutations in the cell-free DNA (cfDNA) of patients with lung cancer and of risk-matched controls reflect clonal haematopoiesis and are non-recurrent. Compared with tumour-derived mutations, clonal haematopoiesis mutations occur on longer cfDNA fragments and lack mutational signatures that are associated with tobacco smoking. Integrating these findings with other molecular features, we develop and prospectively validate a machine-learning method termed 'lung cancer likelihood in plasma' (Lung-CLiP), which can robustly discriminate early-stage lung cancer patients from risk-matched controls. This approach achieves performance similar to that of tumour-informed ctDNA detection and enables tuning of assay specificity in order to facilitate distinct clinical applications. Our findings establish the potential of cfDNA for lung cancer screening and highlight the importance of risk-matching cases and controls in cfDNA-based screening studies.- Published
- 2020
- Full Text
- View/download PDF
43. Deregulation of ribosomal protein expression and translation promotes breast cancer metastasis.
- Author
-
Ebright RY, Lee S, Wittner BS, Niederhoffer KL, Nicholson BT, Bardia A, Truesdell S, Wiley DF, Wesley B, Li S, Mai A, Aceto N, Vincent-Jordan N, Szabolcs A, Chirn B, Kreuzer J, Comaills V, Kalinich M, Haas W, Ting DT, Toner M, Vasudevan S, Haber DA, Maheswaran S, and Micalizzi DS
- Subjects
- Animals, Breast Neoplasms genetics, CRISPR-Cas Systems, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Neoplasm Transplantation, Sequence Analysis, RNA, Breast Neoplasms pathology, Neoplasm Metastasis, Neoplastic Cells, Circulating pathology, Ribosomal Proteins genetics
- Abstract
Circulating tumor cells (CTCs) are shed into the bloodstream from primary tumors, but only a small subset of these cells generates metastases. We conducted an in vivo genome-wide CRISPR activation screen in CTCs from breast cancer patients to identify genes that promote distant metastasis in mice. Genes coding for ribosomal proteins and regulators of translation were enriched in this screen. Overexpression of RPL15 , which encodes a component of the large ribosomal subunit, increased metastatic growth in multiple organs and selectively enhanced translation of other ribosomal proteins and cell cycle regulators. RNA sequencing of freshly isolated CTCs from breast cancer patients revealed a subset with strong ribosome and protein synthesis signatures; these CTCs expressed proliferation and epithelial markers and correlated with poor clinical outcome. Therapies targeting this aggressive subset of CTCs may merit exploration as potential suppressors of metastatic progression., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
44. A post-transcriptional program of chemoresistance by AU-rich elements and TTP in quiescent leukemic cells.
- Author
-
Lee S, Micalizzi D, Truesdell SS, Bukhari SIA, Boukhali M, Lombardi-Story J, Kato Y, Choo MK, Dey-Guha I, Ji F, Nicholson BT, Myers DT, Lee D, Mazzola MA, Raheja R, Langenbucher A, Haradhvala NJ, Lawrence MS, Gandhi R, Tiedje C, Diaz-Muñoz MD, Sweetser DA, Sadreyev R, Sykes D, Haas W, Haber DA, Maheswaran S, and Vasudevan S
- Subjects
- Animals, Cell Cycle, Cells, Cultured, Dual Specificity Phosphatase 1 genetics, Dual Specificity Phosphatase 1 metabolism, Hep G2 Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, K562 Cells, MCF-7 Cells, Mice, Mice, Inbred C57BL, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proteome genetics, Proteome metabolism, THP-1 Cells, Transcriptome, Tristetraprolin genetics, Tumor Necrosis Factor-alpha metabolism, p38 Mitogen-Activated Protein Kinases genetics, p38 Mitogen-Activated Protein Kinases metabolism, AU Rich Elements, Drug Resistance, Neoplasm, RNA Processing, Post-Transcriptional, Tristetraprolin metabolism
- Abstract
Background: Quiescence (G0) is a transient, cell cycle-arrested state. By entering G0, cancer cells survive unfavorable conditions such as chemotherapy and cause relapse. While G0 cells have been studied at the transcriptome level, how post-transcriptional regulation contributes to their chemoresistance remains unknown., Results: We induce chemoresistant and G0 leukemic cells by serum starvation or chemotherapy treatment. To study post-transcriptional regulation in G0 leukemic cells, we systematically analyzed their transcriptome, translatome, and proteome. We find that our resistant G0 cells recapitulate gene expression profiles of in vivo chemoresistant leukemic and G0 models. In G0 cells, canonical translation initiation is inhibited; yet we find that inflammatory genes are highly translated, indicating alternative post-transcriptional regulation. Importantly, AU-rich elements (AREs) are significantly enriched in the upregulated G0 translatome and transcriptome. Mechanistically, we find the stress-responsive p38 MAPK-MK2 signaling pathway stabilizes ARE mRNAs by phosphorylation and inactivation of mRNA decay factor, Tristetraprolin (TTP) in G0. This permits expression of ARE mRNAs that promote chemoresistance. Conversely, inhibition of TTP phosphorylation by p38 MAPK inhibitors and non-phosphorylatable TTP mutant decreases ARE-bearing TNFα and DUSP1 mRNAs and sensitizes leukemic cells to chemotherapy. Furthermore, co-inhibiting p38 MAPK and TNFα prior to or along with chemotherapy substantially reduces chemoresistance in primary leukemic cells ex vivo and in vivo., Conclusions: These studies uncover post-transcriptional regulation underlying chemoresistance in leukemia. Our data reveal the p38 MAPK-MK2-TTP axis as a key regulator of expression of ARE-bearing mRNAs that promote chemoresistance. By disrupting this pathway, we develop an effective combination therapy against chemosurvival.
- Published
- 2020
- Full Text
- View/download PDF
45. Microfluidic concentration and separation of circulating tumor cell clusters from large blood volumes.
- Author
-
Edd JF, Mishra A, Dubash TD, Herrera S, Mohammad R, Williams EK, Hong X, Mutlu BR, Walsh JR, Machado de Carvalho F, Aldikacti B, Nieman LT, Stott SL, Kapur R, Maheswaran S, Haber DA, and Toner M
- Subjects
- Healthy Volunteers, Humans, Lab-On-A-Chip Devices, Microfluidic Analytical Techniques instrumentation, Neoplastic Cells, Circulating pathology
- Abstract
Circulating tumor cells (CTCs) are extremely rare in the blood, yet they account for metastasis. Notably, it was reported that CTC clusters (CTCCs) can be 50-100 times more metastatic than single CTCs, making them particularly salient as a liquid biopsy target. Yet they can split apart and are even rarer, complicating their recovery. Isolation by filtration risks loss when clusters squeeze through filter pores over time, and release of captured clusters can be difficult. Deterministic lateral displacement is continuous but requires channels not much larger than clusters, leading to clogging. Spiral inertial focusing requires large blood dilution factors (or lysis). Here, we report a microfluidic chip that continuously isolates untouched CTC clusters from large volumes of minimally (or undiluted) whole blood. An array of 100 μm-wide channels first concentrates clusters in the blood, and then a similar array transfers them into a small volume of buffer. The microscope-slide-sized PDMS device isolates individually-spiked CTC clusters from >30 mL per hour of whole blood with 80% efficiency into enumeration (fluorescence imaging), and on-chip yield approaches 100% (high speed video). Median blood cell removal (in base-10 logs) is 4.2 for leukocytes, 5.5 for red blood cells, and 4.9 for platelets, leaving less than 0.01% of leukocytes alongside CTC clusters in the product. We also demonstrate that cluster configurations are preserved. Gentle, high throughput concentration and separation of circulating tumor cell clusters from large blood volumes will enable cluster-specific diagnostics and speed the generation of patient-specific CTC cluster lines.
- Published
- 2020
- Full Text
- View/download PDF
46. Quantitative Analysis of Circulating Tumor Cells Using RNA-Based Digital Scoring.
- Author
-
Kalinich M, Kwan TT, Toner M, Haber DA, and Maheswaran S
- Subjects
- Disease Progression, Humans, Neoplasms diagnosis, RNA, Neoplasm genetics, Cell Separation methods, Neoplasms genetics, Neoplasms pathology, Neoplastic Cells, Circulating metabolism, RNA, Neoplasm analysis
- Abstract
Circulating tumor cells (CTCs) provide valuable information about the molecular evolution of cancers, as they may initially respond and ultimately progress on therapy. As intact tumor cells isolated from the bloodstream, CTCs also enable assessment of heterogeneous subpopulations, and their analysis may include DNA, RNA, and protein biomarkers. New microfluidic cell isolation strategies greatly facilitate the challenge of enriching viable tumor cells from the billions of hematopoietic cells within a standard blood specimen. While counting and characterization of enriched CTCs have primarily relied on immunostaining for tumor cell-specific antigens, new RNA-based analytic platforms are providing new insight into the identity of CTCs and providing new tools for clinical applications. Single-cell RNA sequencing of CTCs reveals a high degree of heterogeneity among cancer cells from a single individual, while new digital RNA-based amplification platforms may now allow high-sensitivity and high-throughput quantitative scoring of CTCs for clinical applications. Here, we focus on transcriptomic analysis of CTCs and its relevance in understanding metastatic cancer progression and in developing diagnostic assays to monitor cancer.
- Published
- 2020
- Full Text
- View/download PDF
47. Epithelial to mesenchymal plasticity and differential response to therapies in pancreatic ductal adenocarcinoma.
- Author
-
Porter RL, Magnus NKC, Thapar V, Morris R, Szabolcs A, Neyaz A, Kulkarni AS, Tai E, Chougule A, Hillis A, Golczer G, Guo H, Yamada T, Kurokawa T, Yashaswini C, Ligorio M, Vo KD, Nieman L, Liss AS, Deshpande V, Lawrence MS, Maheswaran S, Fernandez-Del Castillo C, Hong TS, Ryan DP, O'Dwyer PJ, Drebin JA, Ferrone CR, Haber DA, and Ting DT
- Abstract
Transcriptional profiling has defined pancreatic ductal adenocarcinoma (PDAC) into distinct subtypes with the majority being classical epithelial (E) or quasi-mesenchymal (QM). Despite clear differences in clinical behavior, growing evidence indicates these subtypes exist on a continuum with features of both subtypes present and suggestive of interconverting cell states. Here, we investigated the impact of different therapies being evaluated in PDAC on the phenotypic spectrum of the E/QM state. We demonstrate using RNA-sequencing and RNA-in situ hybridization (RNA-ISH) that FOLFIRINOX combination chemotherapy induces a common shift of both E and QM PDAC toward a more QM state in cell lines and patient tumors. In contrast, Vitamin D, another drug under clinical investigation in PDAC, induces distinct transcriptional responses in each PDAC subtype, with augmentation of the baseline E and QM state. Importantly, this translates to functional changes that increase metastatic propensity in QM PDAC, but decrease dissemination in E PDAC in vivo models. These data exemplify the importance of both the initial E/QM subtype and the plasticity of E/QM states in PDAC in influencing response to therapy, which highlights their relevance in guiding clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
48. Modeling gamma radiation exposure rates using geologic and remote sensing data to locate radiogenic anomalies.
- Author
-
Adcock CT, Haber DA, Burnley PC, Malchow RL, and Hausrath EM
- Subjects
- Background Radiation, Gamma Rays, Radiation Exposure statistics & numerical data, Radiation Monitoring, Remote Sensing Technology
- Abstract
Aerial Gamma-Ray Surveys (GRS) are ideal for tracking anthropogenic gamma radiation releases and transport. The interpretation of a GRS can be complicated by natural gamma-ray sources such as atmospheric radon, cosmic rays, geologic materials, and even the survey equipment itself. Some of these complicating factors can be accounted for or corrected by calibration or mathematic techniques. Real-time algorithms that attempt to enhance potential radiogenic anomalies over background are also in use. However, natural geology is a source of significant background gamma-ray production and neither mathematical corrections nor real-time algorithmic approaches directly account for geology and geochemistry. In this study, we advance techniques to predict geologic background exposure rates using rapid and practical methods which can be achieved in the field. In addition we generate models that focus specifically on highlighting radiogenic anomalies for emergency response or further investigation. Predictive models developed in this study were generally able to predict background with medians of ± 1.0 μR/h compared to measured data, and were also able to highlight anomalous areas even where radiation exposure rates were within the range of natural background., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
49. Combination Olaparib and Temozolomide in Relapsed Small-Cell Lung Cancer.
- Author
-
Farago AF, Yeap BY, Stanzione M, Hung YP, Heist RS, Marcoux JP, Zhong J, Rangachari D, Barbie DA, Phat S, Myers DT, Morris R, Kem M, Dubash TD, Kennedy EA, Digumarthy SR, Sequist LV, Hata AN, Maheswaran S, Haber DA, Lawrence MS, Shaw AT, Mino-Kenudson M, Dyson NJ, and Drapkin BJ
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biomarkers, Tumor, Computational Biology methods, Drug Resistance, Neoplasm, Female, Humans, Lung Neoplasms etiology, Lung Neoplasms mortality, Male, Mice, Middle Aged, Neoplasm Recurrence, Local drug therapy, Phthalazines administration & dosage, Piperazines administration & dosage, Small Cell Lung Carcinoma etiology, Small Cell Lung Carcinoma mortality, Temozolomide administration & dosage, Transcriptome, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma pathology
- Abstract
Small-cell lung cancer (SCLC) is an aggressive malignancy in which inhibitors of PARP have modest single-agent activity. We performed a phase I/II trial of combination olaparib tablets and temozolomide (OT) in patients with previously treated SCLC. We established a recommended phase II dose of olaparib 200 mg orally twice daily with temozolomide 75 mg/m
2 daily, both on days 1 to 7 of a 21-day cycle, and expanded to a total of 50 patients. The confirmed overall response rate was 41.7% (20/48 evaluable); median progression-free survival was 4.2 months [95% confidence interval (CI), 2.8-5.7]; and median overall survival was 8.5 months (95% CI, 5.1-11.3). Patient-derived xenografts (PDX) from trial patients recapitulated clinical OT responses, enabling a 32-PDX coclinical trial. This revealed a correlation between low basal expression of inflammatory-response genes and cross-resistance to both OT and standard first-line chemotherapy (etoposide/platinum). These results demonstrate a promising new therapeutic strategy in SCLC and uncover a molecular signature of those tumors most likely to respond. SIGNIFICANCE: We demonstrate substantial clinical activity of combination olaparib/temozolomide in relapsed SCLC, revealing a promising new therapeutic strategy for this highly recalcitrant malignancy. Through an integrated coclinical trial in PDXs, we then identify a molecular signature predictive of response to OT, and describe the common molecular features of cross-resistant SCLC. See related commentary by Pacheco and Byers, p. 1340 . This article is highlighted in the In This Issue feature, p. 1325 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
50. Blood-based monitoring identifies acquired and targetable driver HER2 mutations in endocrine-resistant metastatic breast cancer.
- Author
-
Medford AJ, Dubash TD, Juric D, Spring L, Niemierko A, Vidula N, Peppercorn J, Isakoff S, Reeves BA, LiCausi JA, Wesley B, Malvarosa G, Yuen M, Wittner BS, Lawrence MS, Iafrate AJ, Ellisen L, Moy B, Toner M, Maheswaran S, Haber DA, and Bardia A
- Abstract
Plasma genotyping identifies potentially actionable mutations at variable mutant allele frequencies, often admixed with multiple subclonal variants, highlighting the need for their clinical and functional validation. We prospectively monitored plasma genotypes in 143 women with endocrine-resistant metastatic breast cancer (MBC), identifying multiple novel mutations including HER2 mutations (8.4%), albeit at different frequencies highlighting clinical heterogeneity. To evaluate functional significance, we established ex vivo culture from circulating tumor cells (CTCs) from a patient with HER2 -mutant MBC, which revealed resistance to multiple targeted therapies including endocrine and CDK 4/6 inhibitors, but high sensitivity to neratinib (IC50: 0.018 μM). Immunoblotting analysis of the HER2 -mutant CTC culture line revealed high levels of HER2 expression at baseline were suppressed by neratinib, which also abrogated downstream signaling, highlighting oncogenic dependency with HER2 mutation. Furthermore, treatment of an index patient with HER2 -mutant MBC with the irreversible HER2 inhibitor neratinib resulted in significant clinical response, with complete molecular resolution of two distinct clonal HER2 mutations, with persistence of other passenger subclones, confirming HER2 alteration as a driver mutation. Thus, driver HER2 mutant alleles that emerge during blood-based monitoring of endocrine-resistant MBC confer novel therapeutic vulnerability, and ex vivo expansion of viable CTCs from the blood circulation may broadly complement plasma-based mutational analysis in MBC., Competing Interests: Competing interestsMGH has filed for patent protection for the CTC-iChip technology. D.J. has served as a Consultant/Advisory Board at Novartis, Genentech, Eisai, Ipsen, and EMD Serono. L.S. has served as a Consultant/Advisory Board at Novartis. S.I. has served as a Consultant/Advisory Board at Abbvie, PharaMar, Genentech/Roche, Myriad Genetics, Hengrui Therapeutics, Puma Biotech, and Immunomedics. B.M. spouse has served as a Consultant/Advisory Board at MOTUS GI. A.J.I has served as a Consultant/Advisory Board at Roche, Chugai, Constellation, and Pfizer, and has ownership interests and intellectual property rights/inventor/patent holder at ArcherDx. A.B. has served as a Consultant/Advisory Board at Novartis, Pfizer, Genentech/Roche, Radius Health, Merck, Spectrum pharma, Immunomedics, Sanofi, Daiichi Pharma, and Taiho Oncology, and has research Grant from Biothernostics (self), Genentech (institution), Novartis (institution), Pfizer (institution), Merck (institution), Sanofi (institution), Radius Health (institution), Immunomedics (institution), Mersana(institution), and Innocrin (institution). All other authors declare no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.