Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(1): 14-16, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181738

ABSTRACT

Small cell lung cancer (SCLC) is a recalcitrant malignancy. Conquering it will require deep insight into its biology. In this issue of Cell, Liu and colleagues describe proteomic and phosphoproteomic landscapes of resected SCLC tumors and illustrate the potential of this knowledge to identify new SCLC vulnerabilities.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Proteomics , Knowledge
2.
Cell ; 177(7): 1903-1914.e14, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31031007

ABSTRACT

Xenograft cell transplantation into immunodeficient mice has become the gold standard for assessing pre-clinical efficacy of cancer drugs, yet direct visualization of single-cell phenotypes is difficult. Here, we report an optically-clear prkdc-/-, il2rga-/- zebrafish that lacks adaptive and natural killer immune cells, can engraft a wide array of human cancers at 37°C, and permits the dynamic visualization of single engrafted cells. For example, photoconversion cell-lineage tracing identified migratory and proliferative cell states in human rhabdomyosarcoma, a pediatric cancer of muscle. Additional experiments identified the preclinical efficacy of combination olaparib PARP inhibitor and temozolomide DNA-damaging agent as an effective therapy for rhabdomyosarcoma and visualized therapeutic responses using a four-color FUCCI cell-cycle fluorescent reporter. These experiments identified that combination treatment arrested rhabdomyosarcoma cells in the G2 cell cycle prior to induction of apoptosis. Finally, patient-derived xenografts could be engrafted into our model, opening new avenues for developing personalized therapeutic approaches in the future.


Subject(s)
Animals, Genetically Modified/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Muscle Neoplasms , Rhabdomyosarcoma , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Female , Heterografts , Humans , K562 Cells , Male , Muscle Neoplasms/drug therapy , Muscle Neoplasms/immunology , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Neoplasm Transplantation , Phthalazines/pharmacology , Piperazines/pharmacology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Temozolomide/pharmacology , Xenograft Model Antitumor Assays , Zebrafish/genetics , Zebrafish/immunology
3.
Mol Cell ; 82(18): 3333-3349.e9, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35981542

ABSTRACT

The interaction of RB with chromatin is key to understanding its molecular functions. Here, for first time, we identify the full spectrum of chromatin-bound RB. Rather than exclusively binding promoters, as is often described, RB targets three fundamentally different types of loci (promoters, enhancers, and insulators), which are largely distinguishable by the mutually exclusive presence of E2F1, c-Jun, and CTCF. While E2F/DP facilitates RB association with promoters, AP-1 recruits RB to enhancers. Although phosphorylation in CDK sites is often portrayed as releasing RB from chromatin, we show that the cell cycle redistributes RB so that it enriches at promoters in G1 and at non-promoter sites in cycling cells. RB-bound promoters include the classic E2F-targets and are similar between lineages, but RB-bound enhancers associate with different categories of genes and vary between cell types. Thus, RB has a well-preserved role controlling E2F in G1, and it targets cell-type-specific enhancers and CTCF sites when cells enter S-phase.


Subject(s)
Chromatin , Retinoblastoma Protein , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Promoter Regions, Genetic , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factor AP-1/genetics
4.
Future Oncol ; 15(3): 231-239, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30362375

ABSTRACT

Lurbinectedin is an inhibitor of active transcription of protein-coding genes, causing DNA-break accumulation, apoptosis and modulation of the tumor microenvironment. Early-phase clinical trials indicate promising activity of lurbinectedin in small-cell lung cancer. Here, we describe the rationale and design of ATLANTIS (NCT02566993), an open-label, randomized, multicenter Phase III study to compare the efficacy of lurbinectedin and doxorubicin combination with standard-of-care chemotherapy, investigator's choice of cyclophosphamide/doxorubicin/vincristine or topotecan, in patients with small-cell lung cancer that has progressed following one line of platinum-based chemotherapy. Patients are randomized in a 1:1 ratio. The primary end point is overall survival and key secondary end points include progression-free survival, best tumor response and duration of response, each assessed by independent review committee.


Subject(s)
Carbolines/administration & dosage , Doxorubicin/administration & dosage , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Small Cell Lung Carcinoma/drug therapy , Tumor Microenvironment/drug effects , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cyclophosphamide/administration & dosage , Female , Humans , Male , Middle Aged , Platinum/administration & dosage , Platinum/adverse effects , Progression-Free Survival , Small Cell Lung Carcinoma/pathology , Topotecan/administration & dosage , Vincristine/administration & dosage
5.
Oncogene ; 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39154123

ABSTRACT

Small cell lung cancer (SCLC) is a recalcitrant cancer of neuroendocrine (NE) origin. Changes in therapeutic approaches against SCLC have been lacking over the decades. Here, we use preclinical models to identify a new therapeutic vulnerability in SCLC consisting of the targetable Jumonji lysine demethylase (KDM) family. We show that Jumonji demethylase inhibitors block malignant growth and that etoposide-resistant SCLC cell lines are particularly sensitive to Jumonji inhibition. Mechanistically, small molecule-mediated inhibition of Jumonji KDMs activates endoplasmic reticulum (ER) stress genes, upregulates ER stress signaling, and triggers apoptotic cell death. Furthermore, Jumonji inhibitors decrease protein levels of SCLC NE markers INSM1 and Secretogranin-3 and of driver transcription factors ASCL1 and NEUROD1. Genetic knockdown of KDM4A, a Jumonji demethylase highly expressed in SCLC and a known regulator of ER stress genes, induces ER stress response genes, decreases INSM1, Secretogranin-3, and NEUROD1 and inhibits proliferation of SCLC in vitro and in vivo. Lastly, we demonstrate that two different small molecule Jumonji KDM inhibitors (pan-inhibitor JIB-04 and KDM4 inhibitor SD70) block the growth of SCLC tumor xenografts in vivo. Our study highlights the translational potential of Jumonji KDM inhibitors against SCLC, a clinically feasible approach in light of recently opened clinical trials evaluating this drug class, and establishes KDM4A as a relevant target across SCLC subtypes.

6.
Nat Commun ; 15(1): 672, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38253555

ABSTRACT

There are few effective treatments for small cell lung cancer (SCLC) underscoring the need for innovative therapeutic approaches. This study focuses on exploiting telomerase, a critical SCLC dependency as a therapeutic target. A prominent characteristic of SCLC is their reliance on telomerase activity, a key enzyme essential for their continuous proliferation. Here we utilize a nucleoside analog, 6-Thio-2'-deoxyguanosine (6TdG) currently in phase II clinical trials, that is preferentially incorporated by telomerase into telomeres leading to telomere dysfunction. Using preclinical mouse and human derived models we find low intermittent doses of 6TdG inhibit tumor growth and reduce metastatic burden. Anti-tumor efficacy correlates with a reduction in a subpopulation of cancer initiating like cells (CICs) identified by their expression of L1CAM/CD133 and highest telomerase activity. 6TdG treatment also leads to activation of innate and adaptive anti-tumor responses. Mechanistically, 6TdG depletes CICs and induces type-I interferon signaling leading to tumor immune visibility by activating tumor cell STING signaling. We also observe increased sensitivity to irradiation after 6TdG treatment in both syngeneic and humanized SCLC xenograft models both of which are dependent on the presence of host immune cells. This study underscores the immune-enhancing and metastasis-reducing effects of 6TdG, employing a range of complementary in vitro and in vivo SCLC preclinical models providing a potential therapeutic approach to SCLC.


Subject(s)
Deoxyguanosine/analogs & derivatives , Lung Neoplasms , Small Cell Lung Carcinoma , Telomerase , Thionucleosides , Humans , Animals , Mice , Small Cell Lung Carcinoma/drug therapy , Lung Neoplasms/drug therapy , Drug Delivery Systems , Telomere
7.
Sci Adv ; 10(3): eadh2579, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38241363

ABSTRACT

Although BRCA1/2 mutations are not commonly found in small cell lung cancer (SCLC), a substantial fraction of SCLC shows clinically relevant response to PARP inhibitors (PARPis). However, the underlying mechanism(s) of PARPi sensitivity in SCLC is poorly understood. We performed quantitative proteomic analyses and identified proteomic changes that signify PARPi responses in SCLC cells. We found that the vulnerability of SCLC to PARPi could be explained by the degradation of lineage-specific oncoproteins (e.g., ASCL1). PARPi-induced activation of the E3 ligase HUWE1 mediated the ubiquitin-proteasome system (UPS)-dependent ASCL1 degradation. Although PARPi induced a general DNA damage response in SCLC cells, this signal generated a cell-specific response in ASCL1 degradation, leading to the identification of HUWE1 expression as a predictive biomarker for PARPi. Combining PARPi with agents targeting these pathways markedly improved therapeutic response in SCLC. The degradation of lineage-specific oncoproteins therefore represents a previously unidentified mechanism for PARPi efficacy in SCLC.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , BRCA1 Protein/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proteomics , BRCA2 Protein/genetics , Oncogene Proteins , Cell Line, Tumor , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics
8.
Cancer Discov ; 14(5): 804-827, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38386926

ABSTRACT

Small cell lung cancer (SCLC) presents as a highly chemosensitive malignancy but acquires cross-resistance after relapse. This transformation is nearly inevitable in patients but has been difficult to capture in laboratory models. Here, we present a preclinical system that recapitulates acquired cross-resistance, developed from 51 patient-derived xenograft (PDX) models. Each model was tested in vivo against three clinical regimens: cisplatin plus etoposide, olaparib plus temozolomide, and topotecan. These drug-response profiles captured hallmark clinical features of SCLC, such as the emergence of treatment-refractory disease after early relapse. For one patient, serial PDX models revealed that cross-resistance was acquired through MYC amplification on extrachromosomal DNA (ecDNA). Genomic and transcriptional profiles of the full PDX panel revealed that MYC paralog amplifications on ecDNAs were recurrent in relapsed cross-resistant SCLC, and this was corroborated in tumor biopsies from relapsed patients. We conclude that ecDNAs with MYC paralogs are recurrent drivers of cross-resistance in SCLC. SIGNIFICANCE: SCLC is initially chemosensitive, but acquired cross-resistance renders this disease refractory to further treatment and ultimately fatal. The genomic drivers of this transformation are unknown. We use a population of PDX models to discover that amplifications of MYC paralogs on ecDNA are recurrent drivers of acquired cross-resistance in SCLC. This article is featured in Selected Articles from This Issue, p. 695.


Subject(s)
Drug Resistance, Neoplasm , Gene Amplification , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Mice , Animals , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
9.
Cancer Cell ; 42(8): 1352-1369.e13, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39029464

ABSTRACT

Small cell lung cancers (SCLCs) are composed of heterogeneous subtypes marked by lineage-specific transcription factors, including ASCL1, NEUROD1, and POU2F3. POU2F3-positive SCLCs, ∼12% of all cases, are uniquely dependent on POU2F3 itself; as such, approaches to attenuate POU2F3 expression may represent new therapeutic opportunities. Here using genome-scale screens for regulators of POU2F3 expression and SCLC proliferation, we define mSWI/SNF complexes as top dependencies specific to POU2F3-positive SCLC. Notably, chemical disruption of mSWI/SNF ATPase activity attenuates proliferation of all POU2F3-positive SCLCs, while disruption of non-canonical BAF (ncBAF) via BRD9 degradation is effective in pure non-neuroendocrine POU2F3-SCLCs. mSWI/SNF targets to and maintains accessibility over gene loci central to POU2F3-mediated gene regulatory networks. Finally, clinical-grade pharmacologic disruption of SMARCA4/2 ATPases and BRD9 decreases POU2F3-SCLC tumor growth and increases survival in vivo. These results demonstrate mSWI/SNF-mediated governance of the POU2F3 oncogenic program and suggest mSWI/SNF inhibition as a therapeutic strategy for POU2F3-positive SCLCs.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms , Small Cell Lung Carcinoma , Transcription Factors , Humans , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , Mice , Cell Line, Tumor , Cell Proliferation , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics
10.
Cancer ; 119(14): 2620-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23625575

ABSTRACT

BACKGROUND: Outcomes for patients with chronic myeloid leukemia (CML) have improved after the advent of tyrosine kinase inhibitors (TKIs), which target the BCR/ABL fusion gene product. Nonetheless, differences in survival persist between age groups. The authors performed a retrospective cohort study using the Surveillance, Epidemiology, and End Results (SEER) database to assess 5-year overall survival (OS) in various patient age groups. METHODS: Patients who had a diagnosis of CML were identified using the SEER 19 registries database. Patients who were included had SEER diagnosis codes for CML not otherwise specified (code 9863) and BCR/ABL-positive CML (code 9875) diagnosed between January 2000 and December 2005. Patients were divided into cohorts based on age at diagnosis: ages 15 to 44 years, 45 to 64 years, 65 to 74 years, and 75 to 84 years. OS was estimated using the Kaplan-Meier method, and Cox regression was used to estimate predictors of patient survival. RESULTS: In total, 5138 patients with a new CML diagnosis were identified. Five-year OS improved for all patients between the years 2000 and 2005. Compared with patients who were diagnosed in 2000, 5-year survival improved among patients ages 15 to 44 years (hazard ratio [HR] for mortality, 0.424; P < .0001), ages 45 to 64 years (HR, 0.716; P = .0315), and ages 65 to 74 years (HR, 0.692; P = .0126); and patients ages 75 to 84 years had an increased 5-year OS rate from 19.2% in 2000 to 36.4% in 2005 (HR, 0.568; P < .0001). CONCLUSIONS: OS at 5 years improved among all patients, including those ages 75 to 84 years, a group with historically poor outcomes. However, older age retained an association with worse survival, suggesting opportunities for further progress.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Cause of Death , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Proportional Hazards Models , Retrospective Studies , SEER Program , United States/epidemiology
11.
Am J Hematol ; 88(8): 642-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23619915

ABSTRACT

Acute myeloid leukemia (AML) is more common and more lethal among patients over the age of 60. Increased body mass index (BMI) has been associated with a higher incidence of various malignancies, including AML. We sought to determine whether patient BMI at the time of AML diagnosis is related to overall survival (OS) among elderly patients. We identified 97 patients with AML diagnosed after the age of 60 and treated with cytarabine-based induction chemotherapy. The median age was 68 years (range 60-87); 52% of patients were male, and our study population was predominantly white (89% of patients). The median OS for all patients was 316 days (95% CI 246-459). The hazard ratio for mortality was increased among patients with a BMI < 25 compared to BMI ≥ 30 (HR 2.14, P = 0.009, 95% CI 1.21-3.77), as well as with older age (HR 1.76, P = 0.015, 95% CI 1.12-2.79) and with secondary versus de novo disease (HR 1.95, P = 0.006, 95% CI 1.21-3.14). After multivariable analysis, we did not find a significant association between OS and other potential confounders such as coronary artery disease or diabetes among these patients. We conclude that increased BMI was independently associated with improved OS among older AML patients at our institution.


Subject(s)
Body Mass Index , Leukemia, Myeloid, Acute/mortality , Age Factors , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/administration & dosage , Coronary Artery Disease/mortality , Coronary Artery Disease/therapy , Cytarabine/administration & dosage , Diabetes Mellitus/mortality , Diabetes Mellitus/therapy , Disease-Free Survival , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , Retrospective Studies , Risk Factors , Survival Rate
12.
bioRxiv ; 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37425738

ABSTRACT

Small cell lung cancer (SCLC) presents as a highly chemosensitive malignancy but acquires cross-resistance after relapse. This transformation is nearly inevitable in patients but has been difficult to capture in laboratory models. Here we present a pre-clinical system that recapitulates acquired cross-resistance in SCLC, developed from 51 patient-derived xenografts (PDXs). Each model was tested for in vivo sensitivity to three clinical regimens: cisplatin plus etoposide, olaparib plus temozolomide, and topotecan. These functional profiles captured hallmark clinical features, such as the emergence of treatment-refractory disease after early relapse. Serially derived PDX models from the same patient revealed that cross-resistance was acquired through a MYC amplification on extrachromosomal DNA (ecDNA). Genomic and transcriptional profiles of the full PDX panel revealed that this was not unique to one patient, as MYC paralog amplifications on ecDNAs were recurrent among cross-resistant models derived from patients after relapse. We conclude that ecDNAs with MYC paralogs are recurrent drivers of cross-resistance in SCLC. SIGNIFICANCE: SCLC is initially chemosensitive, but acquired cross-resistance renders this disease refractory to further treatment and ultimately fatal. The genomic drivers of this transformation are unknown. We use a population of PDX models to discover that amplifications of MYC paralogs on ecDNA are recurrent drivers of acquired cross-resistance in SCLC.

13.
Cancer Cell ; 41(9): 1535-1540, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37699331

ABSTRACT

The understanding of small cell lung cancer (SCLC) biology has increased dramatically in recent years, but the processes that allow SCLC to progress rapidly remain poorly understood. Here, we advocate the integration of rapid autopsies and preclinical models into SCLC research as a comprehensive strategy with the potential to revolutionize current treatment paradigms.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Autopsy , Small Cell Lung Carcinoma/genetics , Lung Neoplasms/genetics
14.
Sci Adv ; 8(19): eabn1229, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35559669

ABSTRACT

In small cell lung cancer (SCLC), acquired resistance to DNA-damaging therapy is challenging to study because rebiopsy is rarely performed. We used patient-derived xenograft models, established before therapy and after progression, to dissect acquired resistance to olaparib plus temozolomide (OT), a promising experimental therapy for relapsed SCLC. These pairs of serial models reveal alterations in both cell cycle kinetics and DNA replication and demonstrate both inter- and intratumoral heterogeneity in mechanisms of resistance. In one model pair, up-regulation of translesion DNA synthesis (TLS) enabled tolerance of OT-induced damage during DNA replication. TLS inhibitors restored sensitivity to OT both in vitro and in vivo, and similar synergistic effects were seen in additional SCLC cell lines. This represents the first described mechanism of acquired resistance to DNA damage in a patient with SCLC and highlights the potential of the serial model approach to investigate and overcome resistance to therapy in SCLC.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Cell Line, Tumor , DNA , DNA Damage , DNA Replication , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phthalazines , Piperazines , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Temozolomide/pharmacology
15.
Cancer Cell ; 39(4): 453-456, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33848475

ABSTRACT

Small-cell lung cancer (SCLC) is initially sensitive to platinum doublet chemotherapy, providing dramatic clinical benefit. Unfortunately, most SCLCs relapse and become resistant to further therapy. In this issue of Cancer Cell, Thomas et al. show that some platinum-resistant SCLCs benefit from combination therapy with topotecan plus the ATR (ataxia telangiectasia-mutated and rad3-related) inhibitor berzosertib.


Subject(s)
Lung Neoplasms , Platinum , Antineoplastic Combined Chemotherapy Protocols , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Platinum/therapeutic use , Topotecan/therapeutic use
16.
Article in English | MEDLINE | ID: mdl-32513672

ABSTRACT

Over the past several years, we have witnessed a resurgence of interest in the biology and therapeutic vulnerabilities of small-cell lung cancer (SCLC). This has been driven in part through the development of a more extensive array of representative models of disease, including a diverse variety of genetically engineered mouse models and human tumor xenografts. Herein, we review recent progress in SCLC model development, and consider some of the particularly active avenues of translational research in SCLC, including interrogation of intratumoral heterogeneity, insights into the cell of origin and oncogenic drivers, mechanisms of chemoresistance, and new therapeutic opportunities including biomarker-directed targeted therapies and immunotherapies. Whereas SCLC remains a highly lethal disease, these new avenues of translational research, bringing together mechanism-based preclinical and clinical research, offer new hope for patients with SCLC.


Subject(s)
Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mutation , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Translational Research, Biomedical , Whole Genome Sequencing
17.
Elife ; 102021 08 04.
Article in English | MEDLINE | ID: mdl-34346867

ABSTRACT

Changes in MAPK signaling allow lung cancer cells to transition between lineages that respond differently to treatment.


Subject(s)
Lung Neoplasms , MAP Kinase Signaling System , Humans , Lung , Lung Neoplasms/genetics
18.
J Exp Med ; 218(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34415995

ABSTRACT

T cell immunotherapies have revolutionized treatment for a subset of cancers. Yet, a major hurdle has been the lack of facile and predicative preclinical animal models that permit dynamic visualization of T cell immune responses at single-cell resolution in vivo. Here, optically clear immunocompromised zebrafish were engrafted with fluorescent-labeled human cancers along with chimeric antigen receptor T (CAR T) cells, bispecific T cell engagers (BiTEs), and antibody peptide epitope conjugates (APECs), allowing real-time single-cell visualization of T cell-based immunotherapies in vivo. This work uncovered important differences in the kinetics of T cell infiltration, tumor cell engagement, and killing between these immunotherapies and established early endpoint analysis to predict therapy responses. We also established EGFR-targeted immunotherapies as a powerful approach to kill rhabdomyosarcoma muscle cancers, providing strong preclinical rationale for assessing a wider array of T cell immunotherapies in this disease.


Subject(s)
Immunotherapy/methods , Rhabdomyosarcoma/therapy , Single-Cell Analysis/methods , Xenograft Model Antitumor Assays/methods , Zebrafish/genetics , Adolescent , Adult , Animals , Animals, Genetically Modified , Child , Child, Preschool , DNA-Binding Proteins/genetics , ErbB Receptors/immunology , Female , Humans , Immunotherapy, Adoptive , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice, Inbred Strains , Phthalazines/pharmacology , Piperazines/pharmacology , Rhabdomyosarcoma/pathology , T-Lymphocytes/immunology , Temozolomide/pharmacology , Tumor Cells, Cultured , Zebrafish Proteins/genetics
19.
Mol Syst Biol ; 5: 328, 2009.
Article in English | MEDLINE | ID: mdl-19920813

ABSTRACT

Cyclin-dependent kinase (Cdk) both promotes mitotic entry (spindle assembly and anaphase) and inhibits mitotic exit (spindle disassembly and cytokinesis), leading to an elegant quantitative hypothesis that a single cyclin oscillation can function as a ratchet to order these events. This ratchet is at the core of a published ODE model for the yeast cell cycle. However, the ratchet model requires appropriate cyclin dose-response thresholds. Here, we test the inhibition of mitotic exit in budding yeast using graded levels of stable mitotic cyclin (Clb2). In opposition to the ratchet model, stable levels of Clb2 introduced dose-dependent delays, rather than hard thresholds, that varied by mitotic exit event. The ensuing cell cycle was highly abnormal, suggesting a novel reason for cyclin degradation. Cdc14 phosphatase antagonizes Clb2-Cdk, and Cdc14 is released from inhibitory nucleolar sequestration independently of stable Clb2. Thus, Cdc14/Clb2 balance may be the appropriate variable for mitotic regulation. Although our results are inconsistent with the aforementioned ODE model, revision of the model to allow Cdc14/Clb2 balance to control mitotic exit corrects these discrepancies, providing theoretical support for our conclusions.


Subject(s)
Cyclin B/metabolism , Mitosis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus/drug effects , Anaphase/drug effects , Biosensing Techniques , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , G1 Phase/drug effects , Histones/metabolism , Mating Factor , Mitosis/drug effects , Models, Genetic , Peptides/pharmacology , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
20.
Cancer Cell ; 38(2): 150-152, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32781039

ABSTRACT

The development and consequences of lineage plasticity during tumorigenesis have remained mysterious due to the limits of single-cell analysis. In this issue of Cancer Cell, LaFave et al. and Marjanovic et al. identify highly plastic subpopulations within lung adenocarcinoma that may underlie intratumoral lineage heterogeneity, metastasis, and acquired resistance to chemotherapy.


Subject(s)
Lung Neoplasms , Adenocarcinoma of Lung/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL