297 results on '"E. De Stanchina"'
Search Results
2. Selective Inhibition of HDAC3 Targets Synthetic Vulnerabilities and Activates Immune Surveillance in Lymphoma
- Author
-
Sattva S. Neelapu, David A. Scheinberg, Saber Tadros, Neeraj Jain, E. De Stanchina, Matthew Durant, Jose Baselga Baselga, Lorena Fontan, Eneda Toska, Oliver Weigert, Cassian Yee, Anas Younes, Ahmet Dogan, Ari Melnick, Aaron Y. Chang, Huan Yang, Anja Mottok, Hsia-Yuan Ying, Loretta J. Nastoupil, Chi-Shuen Chu, Stefan Alig, Robert G. Roeder, Giorgio Inghirami, Patrizia Mondello, Man Chun John Ma, Shailbala Singh, Michael R. Green, Sreejoyee Ghosh, and Matt Teater
- Subjects
0301 basic medicine ,Lymphoma ,Genes, MHC Class I ,medicine.disease_cause ,B7-H1 Antigen ,Epigenome ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Immune Checkpoint Inhibitors ,Histone Acetyltransferases ,Mutation ,biology ,CREBBP ,BCL6 ,CREB-Binding Protein ,Oncology ,030220 oncology & carcinogenesis ,Proto-Oncogene Proteins c-bcl-6 ,Interferon ,Growth inhibition ,Epigenetic therapy ,Signal Transduction ,Cyclin-Dependent Kinase Inhibitor p21 ,Antigen-Presenting Cells ,Histone Deacetylases ,Article ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Immune system ,Cell Line, Tumor ,CREBBP, HDAC3, Interferon, MHC class II ,MHC class I ,medicine ,Animals ,Humans ,Cell Proliferation ,Histocompatibility Antigens Class II ,HDAC3 ,Histone acetyltransferase ,Histone Deacetylase Inhibitors ,030104 developmental biology ,chemistry ,Immune System ,MHC class II ,Cancer research ,biology.protein ,Interferons - Abstract
CREBBP mutations are highly recurrent in B-cell lymphomas and either inactivate its histone acetyltransferase (HAT) domain or truncate the protein. Herein, we show that these two classes of mutations yield different degrees of disruption of the epigenome, with HAT mutations being more severe and associated with inferior clinical outcome. Genes perturbed by CREBBP mutation are direct targets of the BCL6–HDAC3 onco-repressor complex. Accordingly, we show that HDAC3-selective inhibitors reverse CREBBP-mutant aberrant epigenetic programming, resulting in: (i) growth inhibition of lymphoma cells through induction of BCL6 target genes such as CDKN1A and (ii) restoration of immune surveillance due to induction of BCL6-repressed IFN pathway and antigen-presenting genes. By reactivating these genes, exposure to HDAC3 inhibitors restored the ability of tumor-infiltrating lymphocytes to kill DLBCL cells in an MHC class I and II–dependent manner, and synergized with PD-L1 blockade in a syngeneic model in vivo. Hence, HDAC3 inhibition represents a novel mechanism-based immune epigenetic therapy for CREBBP-mutant lymphomas. Significance: We have leveraged the molecular characterization of different types of CREBBP mutations to define a rational approach for targeting these mutations through selective inhibition of HDAC3. This represents an attractive therapeutic avenue for targeting synthetic vulnerabilities in CREBBP-mutant cells in tandem with promoting antitumor immunity. This article is highlighted in the In This Issue feature, p. 327
- Published
- 2020
- Full Text
- View/download PDF
3. ERα-LBD, a novel isoform of estrogen receptor alpha, promotes breast cancer proliferation and endocrine resistance
- Author
-
Qing Chang, M. Turkekul, E. Spisni, Brian Houck-Loomis, Chiara Mastroleo, F. Borsetti, Fresia Pareja, David Lyden, Marjan Berishaj, M.F. Berger, Bo Liu, David N Brown, R. Segu Rajappachetty, Fanli Meng, Alexander V Penson, Jorge S. Reis-Filho, S Chandarlapaty, Jacqueline Bromberg, Pasquale Sansone, V. Boyko, A. Strillacci, E. De Stanchina, Ronald C. Hendrickson, and V. R. Rajasekhar
- Subjects
Gene isoform ,Fulvestrant ,Cell growth ,DNA-binding domain ,Biology ,medicine.disease_cause ,medicine.disease ,Metastatic breast cancer ,Breast cancer ,medicine ,Cancer research ,Carcinogenesis ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Estrogen receptor alpha (ERα) drives mammary gland development and breast cancer (BC) growth through an evolutionarily conserved linkage of DNA binding and hormone activation functions. Therapeutic targeting of the hormone binding pocket is a widely utilized and successful strategy for breast cancer prevention and treatment. However, resistance to this endocrine therapy is frequently encountered and may occur through bypass or reactivation of ER-regulated transcriptional programs. We now identify the induction of a novel ERα isoform, ERα-LBD, that is encoded by an alternative ESR1 transcript and lacks the activation function and DNA binding domains. Despite lacking the transcriptional activity, ERα-LBD is found to promote breast cancer growth and resistance to the ERα antagonist fulvestrant. ERα-LBD is predominantly localized to the cytoplasm and mitochondria of BC cells and leads to enhanced glycolysis, respiration and stem-like features. Intriguingly, ERα-LBD expression and function does not appear to be restricted to cancers that express full length ERα but also promotes growth of triple negative breast cancers and ERα-LBD transcript (ESR1-LBD) is also present in BC samples from both ERα(+) and ERα(−) human tumors. These findings point to ERα-LBD as a potential mediator of breast cancer progression and therapy resistance.SIGNIFICANCE STATEMENTEndocrine resistant and metastatic breast cancer (BC) is a clinically significant problem. Our study of fulvestrant resistant cancer cells led to the discovery of a novel ERα isoform which we call ERα-LBD. Encoded by a truncated transcript variant (ESR1-LBD) and lacking the N-terminal domains (activation of transcription and DNA binding), ERα-LBD displays a unique role in BC tumorigenesis and progression by mechanisms that may involve metabolic and cell growth advantages, stemness and therapy resistance. Importantly, ESR1-LBD is preferentially expressed in human breast tumor tissues and may be used as prognostic marker in BC.
- Published
- 2021
- Full Text
- View/download PDF
4. Regional and clonal T cell dynamics at single cell resolution in immune checkpoint blockade
- Author
-
Charles M. Rudin, Daniel K. Wells, Taha Merghoub, P. Manoj, Triparna Sen, H. J. Woo, Marissa Mattar, Jennifer L. Sauter, Joy A. Pai, Hira Rizvi, Mark T.A. Donoghue, Viola Allaj, Matthew D. Hellmann, Ansuman T. Satpathy, Jamie E. Chaft, Nisargbhai S. Shah, Helen Won, Andrew J. Plodkowski, Marina K. Baine, E. De Stanchina, Fathema Uddin, Brian Houck-Loomis, Álvaro Quintanal-Villalonga, Jedd D. Wolchok, Andrew Chow, and Joseph M. Chan
- Subjects
medicine.anatomical_structure ,Single cell sequencing ,T cell ,T-cell receptor ,Cell ,Cancer cell ,Cancer research ,medicine ,Cytotoxic T cell ,Biology ,CD8 ,Immune checkpoint - Abstract
Paired T cell receptor and RNA single cell sequencing (scTCR/RNA-seq) has allowed for enhanced resolution of clonal T cell dynamics in cancer. Here, we report a scTCR/RNA-seq dataset of 162,062 single T cells from 31 tissue regions, including tumor, adjacent normal tissues, and lymph nodes (LN), from three patients who underwent resections for progressing lung cancers after immune checkpoint blockade (ICB). We found marked regional heterogeneity in tumor persistence that was associated with heterogeneity in CD4 and CD8 T cell phenotypes; regions with persistent cancer cells were enriched for follicular helper CD4 T cells (TFH), regulatory T cells (Treg), and exhausted CD8 T cells. Clonal analysis demonstrated that highly-expanded T cell clones were predominantly of the CD8 subtype, were ubiquitously present across all sampled regions, found in the peripheral circulation, and expressed gene signatures of ‘large’ and ‘dual-expanded’ clones that have been predictive of response to ICB. Longitudinal tracking of CD8 T cell clones in the peripheral blood revealed that the persistence of ubiquitous CD8 T cell clones, as well as phenotypically distinct clones with tumor-reactive features, correlated with systemic tumor control. Finally, tracking CD8 T cell clones across tissues revealed the presence of TCF-1+precursor exhausted CD8 T cells in tumor draining LNs that were clonally linked to expanded exhausted CD8 T cells in tumors. Altogether, this comprehensive scTCR/RNA-seq dataset with regional, longitudinal, and clonal resolution provides fundamental insights into the tissue distribution, persistence, and differentiation trajectories of ICB-responsive T cells that underlie clinical responses to ICB.
- Published
- 2021
- Full Text
- View/download PDF
5. Vepafestinib is effective in preclinical models of sarcomas with RET fusion including a brain metastasis model
- Author
-
I. Odintsov, A. Liu, I. Khodos, Q. Chang, C. Giuliano, M. Mattar, M. Vojnic, A. Bonifacio, E. De Stanchina, E. Lovati, M. Ladanyi, and R. Somwar
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
6. 125P LSD1 inhibition restores MHC class I expression and augments the anti-tumor response of immune checkpoint blockade in small cell lung cancer
- Author
-
T. Sen, M.N. Nguyen, H. Taniguchi, A. Chow, E. De Stanchina, and C.M. Rudin
- Subjects
Oncology ,Hematology - Published
- 2021
- Full Text
- View/download PDF
7. MA16.03 CRISPR Screen Reveals XPO1 as a Therapeutic Target Strongly Sensitizing to First and Second Line Therapy in Small Cell Lung Cancer
- Author
-
Yingqian Zhan, Joseph M. Chan, E. De Stanchina, Fathema Uddin, Andrew Chow, Jacklynn V. Egger, U. Bhanot, Viola Allaj, Álvaro Quintanal-Villalonga, Triparna Sen, John T. Poirier, Michael Offin, Yuan Hao, Hirokazu Taniguchi, P. Manoj, Shweta S. Chavan, Charles M. Rudin, J. Qiu, and Nisargbhai S. Shah
- Subjects
Pulmonary and Respiratory Medicine ,Second-line therapy ,XPO1 ,Oncology ,business.industry ,Cancer research ,Medicine ,CRISPR ,Non small cell ,business - Published
- 2021
- Full Text
- View/download PDF
8. 1MO Multi-omic characterization of lung tumors identify AKT and EZH2 as potential therapeutic targets in adenocarcinoma-to-squamous transdifferentiation
- Author
-
Yingqian Zhan, Nisargbhai S. Shah, J. Qiu, P. Manoj, Natasha Rekhtman, Hiroya Taniguchi, Viola Allaj, Metamia Ciampricotti, U. Bhanot, Charles M. Rudin, Brian Houck-Loomis, Maysun Hasan, Jacklynn V. Egger, Álvaro Quintanal-Villalonga, E. De Stanchina, Fathema Uddin, Richard Koche, Shweta S Chavan, Helena Alexandra Yu, and Triparna Sen
- Subjects
Lung ,business.industry ,EZH2 ,Transdifferentiation ,Hematology ,medicine.disease ,Omics ,medicine.anatomical_structure ,Oncology ,Cancer research ,Medicine ,Adenocarcinoma ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
9. MA11.06 Multi-Omic Characterization of Lung Tumors Implicates AKT and MYC Signaling in Adenocarcinoma to Squamous Cell Transdifferentiation
- Author
-
Charles M. Rudin, Helena Alexandra Yu, Jason C. Chang, Joseph M. Chan, Maysun Hasan, Brian Houck-Loomis, Andrew Chow, Álvaro Quintanal-Villalonga, Richard Koche, Triparna Sen, Jacklynn V. Egger, Yingqian Zhan, J. Qiu, M. Offin, P. Manoj, U. Bhanot, E. De Stanchina, Fathema Uddin, Viola Allaj, Nisargbhai S. Shah, Shweta S Chavan, Natasha Rekhtman, and Hiroya Taniguchi
- Subjects
Pulmonary and Respiratory Medicine ,Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,Cell Transdifferentiation ,medicine ,Cancer research ,Adenocarcinoma ,medicine.disease ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
10. 2MO XPO1 inhibition strongly sensitizes to first-line and second-line therapy in small cell lung cancer
- Author
-
Travis J. Hollmann, E. De Stanchina, Fathema Uddin, Metamia Ciampricotti, Triparna Sen, Y. Hao, Jacklynn V. Egger, J. Qiu, Yingqian Zhan, John T. Poirier, Hirokazu Taniguchi, Richard Koche, Andrew Chow, Álvaro Quintanal-Villalonga, Viola Allaj, Joseph M. Chan, Michael Offin, Nisargbhai S. Shah, Charles M. Rudin, and P. Manoj
- Subjects
XPO1 ,Second-line therapy ,Oncology ,business.industry ,First line ,Cancer research ,Medicine ,Hematology ,Non small cell ,business - Published
- 2021
- Full Text
- View/download PDF
11. 1800O Multi-omic characterization of lung tumors implicates AKT and MYC signaling in adenocarcinoma to squamous cell transdifferentiation
- Author
-
Metamia Ciampricotti, Triparna Sen, Maysun Hasan, Jacklynn V. Egger, P. Manoj, Viola Allaj, Yingqian Zhan, Helena Alexandra Yu, E. De Stanchina, J. Qiu, Fathema Uddin, Joseph M. Chan, Nisargbhai S. Shah, Charles M. Rudin, Natasha Rekhtman, Shweta S. Chavan, U. Bhanot, Andrew Chow, Álvaro Quintanal-Villalonga, and Hirokazu Taniguchi
- Subjects
Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,Cell Transdifferentiation ,medicine ,Cancer research ,Adenocarcinoma ,Hematology ,medicine.disease ,business ,Protein kinase B - Published
- 2021
- Full Text
- View/download PDF
12. Obstruction of BRAFV600E transcription by complementary PNA oligomers as a means to inhibit BRAF-mutant melanoma growth
- Author
-
Shyamprasad Deraje Vasudeva, O Surriga, E. De Stanchina, J H Rothman, and Gary K. Schwartz
- Subjects
Cancer Research ,Peptide nucleic acid ,Cell growth ,Mutant ,Virology ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Terminal deoxynucleotidyl transferase ,chemistry ,Transcription (biology) ,030220 oncology & carcinogenesis ,Molecular Medicine ,030212 general & internal medicine ,Strand invasion ,Molecular Biology ,Gene ,DNA - Abstract
Peptide nucleic acid (PNA) oligomers are DNA mimics, which are capable of binding gene sequences 1000-fold more avidly than complementary native DNA by strand invasion and effectively obstruct transcription. Irreversibly obstructing the transcription or replication of a gene sequence, such as BRAFV600E, offers a potential route to specifically target the cancer cell itself. We have employed PNA oligomers to target BRAFV600E in a sequence-specific complementary manner. These PNAs have been modified by appending configurationally stabilizing cationic peptides in order to improve their cellular delivery and target avidity. Our results indicate that exposure of the melanoma cell lines to a modified PNA-peptide conjugate complementary to BRAFV600E mutation sequence results in a concentration-dependent and time-dependent inhibition of cell growth that is specific for the BRAFV600E-mutant melanoma cell lines with inhibition of mRNA and protein expression. Xenograft mouse trials show increased tumor growth delay and necrosis with the BRAFV600E-complementary PNA-peptide conjugates as compared with the saline and scrambled PNA sequence controls. Similarly, quantitative measurement shows a 2.5-fold decrease in Ki67 and a 3-fold increase in terminal deoxynucleotidyl transferase dUTP nick end labeling expression with this approach. PNA-delivery peptide conjugates represent a novel way to target BRAFV600E and represent a new approach in targeting selective oncogenes that induce tumor growth.
- Published
- 2017
- Full Text
- View/download PDF
13. The anti-HER3 monoclonal antibody seribantumab effectively inhibits growth of patient-derived and isogenic cell line and xenograft models with NRG1 rearrangements
- Author
-
W.J. Sisso, Igor Odintsov, Lukas Delasos, Marissa Mattar, Eric Gladstone, E.M. Sisso, Shawn M. Leland, M. Ladanyi, Morana Vojnic, D. Plessinger, Inna Khodos, Romel Somwar, E. De Stanchina, and Allan Jo-Weng Lui
- Subjects
Cancer Research ,Oncology ,Chemistry ,medicine.drug_class ,Cell culture ,medicine ,Seribantumab ,Monoclonal antibody ,Molecular biology - Published
- 2020
- Full Text
- View/download PDF
14. Selective inhibition of HDAC3 targets synthetic vulnerabilities and activates immune surveillance in lymphoma
- Author
-
Sattva S. Neelapu, Matt Teater, Michael R. Green, Matthew Durant, David A. Scheinberg, Saber Tadros, Ahmet Dogan, E. De Stanchina, Stefan Alig, Oliver Weigert, Giorgio Inghirami, Patrizia Mondello, Anas Younes, Anja Mottok, Cassian Yee, Aaron Y. Chang, Jose Baselga Baselga, Shailbala Singh, Eneda Toska, Lorena Fontan, Neeraj Jain, Ari Melnick, Huan Yang, Man Chun John Ma, and Loretta J. Nastoupil
- Subjects
0303 health sciences ,Mutation ,biology ,Antigen presentation ,Mutant ,Epigenome ,Histone acetyltransferase ,BCL6 ,HDAC3 ,medicine.disease_cause ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,MHC class I ,biology.protein ,medicine ,Cancer research ,030304 developmental biology - Abstract
CREBBP mutations are highly recurrent in B-cell lymphomas and either inactivate its histone acetyltransferase (HAT) domain or truncate the protein. Herein, we show that these two classes of mutations yield different degrees of disruption of the epigenome, with HAT mutations being more severe and associated with inferior clinical outcome. Genes perturbed by CREBBP mutation are direct targets of the BCL6/HDAC3 onco-repressor complex. Accordingly, we show that HDAC3 selective inhibitors fully reverse CREBBP mutant aberrant epigenetic programming resulting in: a) growth inhibition of lymphoma cells through induction of BCL6 target genes such as CDKN1A and b) restoration of immune surveillance due to induction of BCL6 repressed IFN pathway and antigen presentation genes. By reactivating these genes, exposure to HDAC3-i restored the ability of tumor infiltrating lymphocytes to kill DLBCL cells in an MHC II and MHC I dependent manner. Hence HDAC3-i represent a novel mechanism-based immune-epigenetic therapy for CREBBP mutant lymphomas.STATEMENT OF SIGNIFICANCEWe have leveraged the molecular characterization of different types of CREBBP mutations to define a rational approach for targeting these mutations through selective inhibition of HDAC3. This represents an attractive therapeutic avenue for targeting synthetic vulnerabilities in CREBBP mutant cells in tandem with promoting anti-tumor immunity.
- Published
- 2019
- Full Text
- View/download PDF
15. Abstract P4-14-24: Optimization of intermittent high dose lapatinib administration with or without capecitabine: A rational approach to drug dosing and scheduling using Norton-Simon modeling
- Author
-
Bob T. Li, Andrew D. Seidman, Larry Norton, S. Patil, S Chandarlapaty, Aki Morikawa, and E De Stanchina
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.drug_class ,Cancer ,Pharmacology ,medicine.disease ,Lapatinib ,Tyrosine-kinase inhibitor ,Capecitabine ,Breast cancer ,Tolerability ,Internal medicine ,Toxicity ,medicine ,Dosing ,skin and connective tissue diseases ,business ,medicine.drug - Abstract
Background: Systemic treatment of central nervous system (CNS) metastases remains a challenge partially due to poor drug penetration. Lapatinib and capecitabine are drugs with modest efficacy in treatment of brain metastases from HER2-positive (+) breast cancer (BC) and were shown to cross the blood-tumor barrier in clinical craniotomy specimens (Lin N et al., CCR 2009, Morikawa A et al., Neuro Oncol 2015). However, intratumoral drug concentrations observed were sub-optimal and heterogeneous. Administration of shorter-duration, high dose tyrosine kinase inhibitor is proposed as a way to improve efficacy and tolerability based on Norton-Simon modeling and drug exposure in the CNS (Traina T et al., JCO 2008, Grommes C et al., Neuro Oncol 2011, Chien AJ et al., J Clin Oncol 2014) . In this study, we examined optimization of high dose lapatinib administration with or without capecitabine to inform the design of a phase I trial for BC patients with HER2+ CNS metastases. Methods: Mice bearing BT-474 BC xenograft tumors were treated with various lapatinib doses and schedules. A standard continuous daily dose (100mg/kg) was compared to various intermittent dosing schedules (at 100mg/kg, 400mg/kg, and 800mg/kg). In addition, high dose lapatinib (800mg/kg) was administered with capecitabine either concurrently or in tandem. Xenografts were treated when tumors reached 100mm3. Tumor volumes were evaluated for antitumor efficacy, and mice weights were measured for toxicity. Significance testing for between-group comparisons was conducted using a mixed effect model for repeated measures. Results: Intermittent schedules of lapatinib at 100mg/kg given as 3 days on/11 days off (3/11), 5 days on/9 days off (5/9), and 7 days on/7days off (7/7) had a similar efficacy in tumor control: percent change in tumor volume of 225% (7/7), 222% (5/9), and 223% (3/11) (NS). Therefore, the 3 days on (with 4 days off or 11 days off ) schedule was subsequently chosen to evaluate for tolerability and antitumor efficacy of higher lapatinib dose. The 3 days on/4 days off (3/4) group at 800mg/kg demonstrated the highest tumor reduction (-69%) compared to the daily continuous dosing group (-18%) (p=0.04), but a trend toward higher toxicity was observed (p=0.12). Evaluation of concurrent vs. tandem administration of capecitabine with lapatinib at 800mg/kg given in 3 days on/11 days off was conducted. The concurrent treatment was discontinued early due to high toxicity. However, tandem administration of capecitabine with high dose lapatinib was tolerable without a significant difference in weight changes (p=0.62). Conclusions: The intermittent schedule allows delivery of high dose lapatinib, which has better anti-tumor activity than standard continuous dosing. If given intermittently, high dose lapatinib is tolerable, even with capecitabine if given in tandem/sequence. Based on the result of these experiments, a phase I trial of high dose lapatinib using 3 days on/11 days off schedule in tandem with capecitabine is currently proposed for treatment of HER2-positive BC patient with CNS metastases. Citation Format: Morikawa A, De Stanchina E, Patil S, Chandarlapaty S, Li BT, Norton L, Seidman AD. Optimization of intermittent high dose lapatinib administration with or without capecitabine: A rational approach to drug dosing and scheduling using Norton-Simon modeling. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P4-14-24.
- Published
- 2016
- Full Text
- View/download PDF
16. P1.12-15 PET Imaging of [18F]PARP Inhibitor as a Pharmacodynamic Biomarker of Talazoparib in Small Cell Lung Cancer PDXs
- Author
-
Brandon Carney, E. De Stanchina, Susanne Kossatz, Benjamin H. Lok, John T. Poirier, James Laird, Charles M. Rudin, and Thomas Reiner
- Subjects
Pulmonary and Respiratory Medicine ,business.industry ,Pet imaging ,chemistry.chemical_compound ,Oncology ,chemistry ,Pharmacodynamics ,PARP inhibitor ,Cancer research ,Medicine ,Biomarker (medicine) ,Talazoparib ,Non small cell ,business - Published
- 2019
- Full Text
- View/download PDF
17. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2
- Author
-
Charles M. Rudin, E. De Stanchina, John T. Poirier, Christine L. Hann, Andre L. Moreira, Nick Connis, and Eric E. Gardner
- Subjects
Cancer Research ,Lung Neoplasms ,Blotting, Western ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Mice ,Genetics ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,EZH2 ,RNA, Messenger ,Promoter Regions, Genetic ,Molecular Biology ,Regulation of gene expression ,epigenetics ,Reverse Transcriptase Polymerase Chain Reaction ,subtypes ,Polycomb Repressive Complex 2 ,Cancer ,SCLC ,Methylation ,Cell cycle ,DNA Methylation ,medicine.disease ,Molecular biology ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,3. Good health ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,CpG site ,Histone methyltransferase ,DNA methylation ,Cancer research ,CpG Islands ,methylation - Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy characterized by early metastasis, rapid development of resistance to chemotherapy, and genetic instability. This study profiles DNA methylation in SCLC, patient-derived xenografts (PDXs) and cell lines at single nucleotide resolution. DNA methylation patterns of primary samples are distinct from those of cell lines, while PDXs maintain a pattern closely consistent with primary samples. Clustering of DNA methylation and gene expression of primary SCLC revealed distinct disease subtypes among histologically indistinguishable primary patient samples with similar genetic alterations. SCLC is notable for dense clustering of high-level methylation in discrete promoter CpG islands, in a pattern clearly distinct from other lung cancers and strongly correlated with high expression of the E2F target and histone methyltransferase gene EZH2. Pharmacologic inhibition of EZH2 in a SCLC PDX markedly inhibited tumor growth.
- Published
- 2015
18. Obstruction of BRAF
- Author
-
J H, Rothman, O, Surriga, E, de Stanchina, S D, Vasudeva, and G K, Schwartz
- Subjects
Peptide Nucleic Acids ,Proto-Oncogene Proteins B-raf ,Mice ,Amino Acid Substitution ,Transcription, Genetic ,Mutation, Missense ,Animals ,Humans ,Mice, Nude ,Female ,Melanoma ,Xenograft Model Antitumor Assays - Abstract
Peptide nucleic acid (PNA) oligomers are DNA mimics, which are capable of binding gene sequences 1000-fold more avidly than complementary native DNA by strand invasion and effectively obstruct transcription. Irreversibly obstructing the transcription or replication of a gene sequence, such as BRAF
- Published
- 2017
19. Human PGBD5 DNA transposase promotes site-specific oncogenic mutations in rhabdoid tumors
- Author
-
Catherine Reeves, Anne-Katrin Emde, Alex Kentsis, Minita Shah, A Eisenberg, E de Stanchina, Stephen P. Jackson, Elias Rodriguez-Fos, CR Antonescu, Kanika Arora, AG Henssen, Scott A. Armstrong, Eric Still, Jiali Zhuang, Mithat Gonen, Andrew N. Blackford, Hanno Steen, Zhiping Weng, Richard Koche, ND Socci, David Torrents, Casie Reed, Elizabeth J. Perlman, Christopher E. Mason, Montserrat Puiggròs, Santiago Gonzalez, Cwm Roberts, Elizabeth Mullen, and E Jiang
- Subjects
0301 basic medicine ,Transposable element ,Genetics ,Somatic cell ,DNA repair ,Rhabdoid tumors ,Biology ,DNA sequencing ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Cancer research ,Ectopic expression ,Gene ,Transposase ,DNA - Abstract
Genomic rearrangements are a hallmark of childhood solid tumors, but their mutational causes remain poorly understood. Here, we identify the piggyBac transposable element derived 5 (PGBD5) gene as an enzymatically active human DNA transposase expressed in the majority of rhabdoid tumors, a lethal childhood cancer. Using assembly-based whole-genome DNA sequencing, we observed previously unknown somatic genomic rearrangements in primary human rhabdoid tumors. These rearrangements were characterized by deletions and inversions involving PGBD5-specific signal (PSS) sequences at their breakpoints, with some recurrently targeting tumor suppressor genes, leading to their inactivation. PGBD5 was found to be physically associated with human genomic PSS sequences that were also sufficient to mediate PGBD5-induced DNA rearrangements in rhabdoid tumor cells. We found that ectopic expression of PGBD5 in primary immortalized human cells was sufficient to promote penetrant cell transformation in vitro and in immunodeficient mice in vivo. This activity required specific catalytic residues in the PGBD5 transposase domain, as well as end-joining DNA repair, and induced distinct structural rearrangements, involving PSS-associated breakpoints, similar to those found in primary human rhabdoid tumors. This defines PGBD5 as an oncogenic mutator and provides a plausible mechanism for site-specific DNA rearrangements in childhood and adult solid tumors.
- Published
- 2017
- Full Text
- View/download PDF
20. Acquisition and Storage of Clinical Samples to Establish PDX Models
- Author
-
Marissa Mattar, E. De Stanchina, and Omar Abdel-Wahab
- Subjects
0301 basic medicine ,Protocol (science) ,business.industry ,Process (engineering) ,Sample (statistics) ,Institutional review board ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Documentation ,030220 oncology & carcinogenesis ,Key (cryptography) ,Medicine ,Operations management ,business - Abstract
Obtaining and propagating patient-derived samples is a complex process that requires a high degree of coordination among a number of different services spanning from the clinic to the research side, and presents challenges that are administrative, procedural, and technical. Patients need to be selected according to specific criteria and consented to the correct institutional review board protocol; surgery and pathology personnel need to be alerted; samples must be retrieved and delivered to research personnel in a timely and organized manner; samples must be skillfully processed and implanted in mice or stored for subsequent analysis; and sample annotation and proper documentation of each step has to be maintained throughout the entire process. In this chapter, we will discuss the key aspects of this intricate process.
- Published
- 2017
- Full Text
- View/download PDF
21. List of Contributors
- Author
-
O. Abdel-Wahab, A. Akcakanat, S.E. Boyle, T. Brabb, C. Brayton, A. Bruna, D.M. Burgenske, J.W. Cassidy, S. Chateau-Joubert, W. Cheng, Y. Chen, L.A. Colby, E. Corwin, E. de Stanchina, O. Duchamp, J. Eswaraka, K.W. Evans, B. Fang, C.G. Fedele, J.B. Fleming, J.-J. Fontaine, A. Giddabasappa, E. Girard, L.R. Hill, D.K. Hirenallur-Shanthappa, G.Y. Ho, G. Inghirami, B.M. Iritani, Y. Jiang, S.D. Kaffenberger, A. Krivtsov, M.G. Lawrence, L. Liang, J. Liu, J.P. MacKeigan, E. Marangoni, M. Mattar, I. Mercado-Uribe, F. Meric-Bernstam, G.B. Mills, N. Niu, J.M. Olson, N. Paez-Arango, K. Paz, K. Pham, S.P.S. Pillai, M. Pizzi, J.T. Poirier, J.A. Ramírez, M.V. Rios Perez, G. Risbridger, P.J. Russell, P. Sathyan, M. Scaltriti, S.C. Schmechel, C. Scott, C.L. Scott, J.-L. Servely, M. Shackleton, B.W. Simons, J. Snyder, A.K. Sood, A.D. Strand, R. Taylor, S. Thompson-Iritani, P.T. Tinkey, R. Uthamanthil, L. Wang, E.D. Williams, E. Yuca, J. Zhang, and R. Zhang
- Published
- 2017
- Full Text
- View/download PDF
22. Patient-Derived Tumor Xenograft
- Author
-
Peggy T. Tinkey, E. De Stanchina, Rajesh Uthamanthil, and Giorgio Inghirami
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Biological Specimen Banks ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Animal model ,030220 oncology & carcinogenesis ,Medicine ,Medical physics ,business ,Tumor xenograft ,Biomedical engineering - Abstract
Patient-derived tumor xenografts have suddenly emerged as a highly desirable platform in oncology. Many applications have been explored and many others are on the horizon, all with the main objective of improving molecular and biological discoveries, executing more effective drug screenings, and providing new patient customized therapies. Although impressive results have already been rapidly achieved, many questions remain and newer applications continue to be explored. In this chapter, we review the most pressing issues and proposed possible solutions. Future challenges and innovative applications have been introduced.
- Published
- 2017
- Full Text
- View/download PDF
23. Methodologies for Developing and Maintaining Patient-Derived Xenograft Mouse Models
- Author
-
E. De Stanchina, Omar Abdel-Wahab, Maurizio Scaltriti, John T. Poirier, and Marissa Mattar
- Subjects
0301 basic medicine ,03 medical and health sciences ,Engineering ,030104 developmental biology ,0302 clinical medicine ,Risk analysis (engineering) ,business.industry ,030220 oncology & carcinogenesis ,business ,Tumor xenograft ,Tumor transplantation ,Biomedical engineering - Abstract
Successful generation and propagation of patient-derived xenograft models requires a supportive infrastructure composed of dedicated laboratory and animal facilities, regulatory approvals, detailed standard operative procedures, and, importantly, skilled technical staff. This chapter will detail the methodologies involved in the processing and implantation of freshly collected tumor samples and in the maintenance and propagation of successfully established PDX models. In addition, we will discuss the critical parameters influencing the likelihood of successful generation of a PDX and how to avoid the most likely pitfalls.
- Published
- 2017
- Full Text
- View/download PDF
24. Running a PDX Core Laboratory or a PDX Support Program
- Author
-
E. De Stanchina, Rajesh Uthamanthil, Marissa Mattar, and Andrei V. Krivtsov
- Subjects
0301 basic medicine ,Engineering ,Process management ,business.industry ,0402 animal and dairy science ,Cost accounting ,04 agricultural and veterinary sciences ,040201 dairy & animal science ,Biobank ,Variety (cybernetics) ,03 medical and health sciences ,030104 developmental biology ,Workflow ,Key (cryptography) ,Core laboratory ,business ,Simulation - Abstract
Patient-derived xenografts (PDXs) are a powerful technology with the potential to unlock key mechanisms of disease for a variety of difficult-to-treat cancers and cancers with no applicable laboratory models. While individual laboratories can excel at establishing such models, there are procedural, technical, and financial challenges which present serious obstacles for established investigators as well as new investigators without prior experience with PDX models. Therefore, for some institutions, it is preferable to designate a centralized “PDX Core” to streamline the workflow that is required for the success of such a program and make it more cost-effective. Here we describe the key infrastructural components of a successful academic PDX program, underscore the need for well-established procedures and standardized protocols, and discuss cost management strategies.
- Published
- 2017
- Full Text
- View/download PDF
25. PARP trapping by Talazoparib is a Potent Mechanism of Radiosensitization in Small Cell Lung Cancer Cell Lines and Patient-Derived Xenografts
- Author
-
Jennifer Ma, Charles M. Rudin, Benjamin H. Lok, Andrew C. Bell, James Laird, John T. Poirier, and E. De Stanchina
- Subjects
0301 basic medicine ,Cancer Research ,Radiation ,Mechanism (biology) ,business.industry ,Poly ADP ribose polymerase ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Talazoparib ,Medicine ,Radiology, Nuclear Medicine and imaging ,Non small cell ,business - Published
- 2018
- Full Text
- View/download PDF
26. MA21.01 Generation and Characterization of Novel Preclinical Disease Models of NSCLC with NRG1 Rearrangements to Improve Therapy
- Author
-
Morana Vojnic, M. Offin, Hiroki Sato, M. Ladanyi, Alison M. Schram, Marissa Mattar, Lukas Delasos, Evan Siau, Ryma Benayed, Allan Jo-Weng Lui, E. De Stanchina, A. Drilon, Robert Michael Daly, Inna Khodos, Eric Gladstone, Romel Somwar, and R. Kurth
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,business.industry ,Cancer research ,Medicine ,Disease ,business - Published
- 2019
- Full Text
- View/download PDF
27. Blocking metastatic behavior of MUC16/CA-125-expressing cancer by targeting galectin-3
- Author
-
E. Smith, Britta Weigelt, T. White, Thapi Dharma Rao, David R. Spriggs, E. De Stanchina, I.C. Lorenz, Noah Z. Feit, and Marina Stasenko
- Subjects
Oncology ,business.industry ,Blocking (radio) ,Galectin-3 ,Cancer research ,Obstetrics and Gynecology ,Medicine ,Cancer ,business ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
28. Abstract PD1-8: Acquired resistance to rapamycin and mTOR kinase inhibitors is mediated by non-overlapping mutations in distinct sites in the mTOR protein
- Author
-
V Rodrik-Outmezguine, Z Yao, M Berger, E De Stanchina, T Klinowska, D Barratt, J Baselga, and N Rosen
- Subjects
Cancer Research ,Oncology - Abstract
The mammalian target of rapamycin (mTOR) protein kinase regulates protein translation and proliferation by integrating the availability of energy, nutrients and the presence of growth factors. Dysregulation of mTOR is a common event in human tumors and results from mutational activation of receptor tyrosine kinases, PI3K signaling or inactivating mutations of the Tuberous Sclerosis or GATOR complexes as well as the LKB/AMP kinase pathway. The prevalence of mTOR activation in cancer has led to the development of two classes of inhibitors of the protein as therapeutics: the natural product rapamycin and its analogs as well as direct inhibitors of mTOR kinase. mTOR exists in at least two multi-protein complexes. The mTORC1 complex phosphorylates S6K and 4EBP and stimulates protein translation, metabolism as well as other processes. The mTORC2 complex phosphorylates and activates AKT and other AGC kinases. Rapamycin binds to the immunophilin FKBP12. Drug-bound FKBP12 complex binds to mTOR FRB domain and selectively inhibits the activity of mTORC1. However, rapalogs preferentially inhibit S6K phosphorylation compared to 4EBP phosphorylation. Rapalogs have undergone extensive clinical testing and have significant antitumor activity in renal cell and pancreatic neuroendocrine tumors and, in combination with aromatase inhibitors, in resistant, ER positive breast cancers. In contrast, mTOR kinase inhibitors suppress both mTORC1 and mTORC2 functions and potently inhibit S6K, 4EBP and AKT S473 phosphorylation. These drugs are in early clinical testing. In order to better understand the mechanism of action of these drugs and potential mechanisms of tumor escape from mTOR inhibition, we selected breast tumor cells for resistance to growth inhibition in cell culture by treatment with either rapamycin or an mTOR kinase inhibitor. In rapamycin resistant cells, phosphorylation of S6K and S6 were insensitive to the drug, but remained sensitive to mTOR kinase inhibitors. Conversely, in clones resistant to mTOR kinase inhibitors, mTORC1 and mTORC2 substrates were insensitive to the drugs, but S6K and S6 phosphorylation remained sensitive to rapamycin. Deep sequencing results explained these findings: rapamycin resistant clones harbored mutations in the FRB domain of mTOR, in the sites shown to interact with the FKBP12-rapamycin complex; mTOR kinase resistant clones harbored mutations in the mTOR catalytic domain. These mutations were not observed in the parental cells. It is likely that the mutations identified in each domain prevent binding of the drug. Consistent with these data, growth of rapamycin resistant cells retain sensitivity to mTOR kinase inhibitors and mTOR kinase resistant cells retain sensitivity to rapalogs, both in tissue culture and in vivo. The results suggest that tumor cells with acquired resistance to mTOR inhibitors retain a requirement for mTOR signaling for proliferation. Furthermore, tumors resistant to either class of drug may not be cross-resistant to the other and combined therapy with both might delay the onset of resistance. Further studies on the genetics of human tumors with acquired resistance to these agents will determine the clinical importance of these findings. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr PD1-8.
- Published
- 2013
- Full Text
- View/download PDF
29. P3.15-008 [F18]PARPi PET as an In Vivo Pharmacodynamic Biomarker of PARP Inhibitor Therapy in Patient-Derived Xenografts of Small Cell Lung Cancer
- Author
-
P. Donabedian, Benjamin H. Lok, Charles M. Rudin, Nisargbhai S. Shah, Thomas Reiner, Susanne Kossatz, John T. Poirier, E. De Stanchina, A. Sadique, James Laird, and Brandon Carney
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,business.industry ,In vivo ,Internal medicine ,Pharmacodynamics ,PARP inhibitor ,medicine ,Biomarker (medicine) ,In patient ,Non small cell ,business - Published
- 2017
- Full Text
- View/download PDF
30. MA22.01 PARP Inhibitor Radiosensitization of Small Cell Lung Cancer Differs by PARP Trapping Potency
- Author
-
James Laird, John T. Poirier, Jennifer Ma, Benjamin H. Lok, E. De Stanchina, and Charles M. Rudin
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,business.industry ,Poly ADP ribose polymerase ,PARP inhibitor ,Cancer research ,Medicine ,Potency ,Non small cell ,business - Published
- 2018
- Full Text
- View/download PDF
31. p53 tumor suppressor protein regulates the levels of huntingtin gene expression
- Author
-
Josephine Hoh, Andrew Zupnick, Shengkan Jin, Arnold J. Levine, Zhaohui Feng, Scott W. Lowe, Carol Prives, and E. de Stanchina
- Subjects
Chromatin Immunoprecipitation ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Time Factors ,Huntingtin ,Transcription, Genetic ,Blotting, Western ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Response Elements ,Mice ,Huntington's disease ,SETD2 ,Cell Line, Tumor ,mental disorders ,Genetics ,medicine ,Huntingtin Protein ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,Transcription factor ,Mice, Knockout ,Regulation of gene expression ,Reverse Transcriptase Polymerase Chain Reaction ,Temperature ,Nuclear Proteins ,Polyglutamine tract ,Blotting, Northern ,medicine.disease ,Molecular biology ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Huntington Disease ,Gamma Rays ,Mutation ,Tumor Suppressor Protein p53 ,Signal transduction ,DNA Damage ,Signal Transduction - Abstract
The p53 protein is a transcription factor that integrates various cellular stress signals. The accumulation of the mutant huntingtin protein with an expanded polyglutamine tract plays a central role in the pathology of human Huntington's disease. We found that the huntingtin gene contains multiple putative p53-responsive elements and p53 binds to these elements both in vivo and in vitro. p53 activation in cultured human cells, either by a temperature-sensitive mutant p53 protein or by gamma-irradiation (gamma-irradiation), increases huntingtin mRNA and protein expression. Similarly, murine huntingtin also contains multiple putative p53-responsive elements and its expression is induced by p53 activation in cultured cells. Moreover, gamma-irradiation, which activates p53, increases huntingtin gene expression in the striatum and cortex of mouse brain, the major pathological sites for Huntington's disease, in p53+/+ but not the isogenic p53-/- mice. These results demonstrate that p53 protein can regulate huntingtin expression at transcriptional level, and suggest that a p53 stress response could be a modulator of the process of Huntington's disease.
- Published
- 2005
- Full Text
- View/download PDF
32. [ 18 F]PARPi PET as a Marker of Intratumoral Talazoparib Level in Small Cell Lung Cancer
- Author
-
Susanne Kossatz, E. De Stanchina, Charles M. Rudin, Thomas Reiner, James Laird, Benjamin H. Lok, Brandon Carney, Nisargbhai S. Shah, John T. Poirier, Jennifer Ma, A. Sadique, and Andrew C. Bell
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Radiation ,business.industry ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,Talazoparib ,Radiology, Nuclear Medicine and imaging ,Non small cell ,business - Published
- 2017
- Full Text
- View/download PDF
33. Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF
- Author
-
Qing-Bai She, E. De Stanchina, Jedd D. Wolchok, Igor Dolgalev, Taha Merghoub, Yogindra Persaud, Barry S. Taylor, David Cobrinik, Alan N. Houghton, Feng Xing, Humilidad F. Gallardo, Gideon Bollag, Manickam Janakiraman, David B. Solit, Adriana Heguy, Cailian Liu, B. Hefter, Agnes Viale, and Christine A. Pratilas
- Subjects
Proto-Oncogene Proteins B-raf ,Cancer Research ,Biology ,medicine.disease_cause ,Retinoblastoma Protein ,Article ,Mice ,CDKN2A ,Genetics ,medicine ,PTEN ,Animals ,Humans ,Protein kinase A ,Molecular Biology ,neoplasms ,Melanoma ,Phosphoinositide-3 Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Mutation ,Oncogene ,MEK inhibitor ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Cyclin-Dependent Kinase 4 ,medicine.disease ,eye diseases ,Cancer research ,biology.protein ,raf Kinases ,Proto-Oncogene Proteins c-akt ,V600E - Abstract
Identifying the spectrum of genetic alterations that cooperate with critical oncogenes to promote transformation provides a foundation for understanding the diversity of clinical phenotypes observed in human cancers. Here, we performed integrated analyses to identify genomic alterations that co-occur with oncogenic BRAF in melanoma and abrogate cellular dependence upon this oncogene. We identified concurrent mutational inactivation of the PTEN and RB1 tumor suppressors as a mechanism for loss of BRAF/MEK dependence in melanomas harboring (V600E)BRAF mutations. RB1 alterations were mutually exclusive with loss of p16(INK4A), suggesting that whereas p16(INK4A) and RB1 may have overlapping roles in preventing tumor formation, tumors with loss of RB1 exhibit diminished dependence upon BRAF signaling for cell proliferation. These findings provide a genetic basis for the heterogeneity of clinical outcomes in patients treated with targeted inhibitors of the mitogen-activated protein kinase pathway. Our results also suggest a need for comprehensive screening for RB1 and PTEN inactivation in patients treated with RAF and MEK-selective inhibitors to determine whether these alterations are associated with diminished clinical benefit in patients whose cancers harbor mutant BRAF.
- Published
- 2011
34. INK4a/ARF mutations accelerate lymphomagenesis and promote chemoresistance by disabling p53
- Author
-
E. de Stanchina, Mila E. McCurrach, Clemens A. Schmitt, Scott W. Lowe, and R. R. Wallace-Brodeur
- Subjects
Male ,Lymphoma, B-Cell ,Transgene ,Drug Resistance ,Genes, myc ,Locus (genetics) ,Antineoplastic Agents ,Apoptosis ,Mice, Transgenic ,Biology ,law.invention ,Mice ,law ,hemic and lymphatic diseases ,Tumor Suppressor Protein p14ARF ,Genetics ,medicine ,Animals ,Humans ,B-cell lymphoma ,Gene ,Mice, Knockout ,Retinoblastoma ,Genes, p16 ,Proteins ,medicine.disease ,Genes, p53 ,Molecular biology ,Lymphoma ,Mice, Inbred C57BL ,Enhancer Elements, Genetic ,Mutation ,Cancer research ,Immunoglobulin heavy chain ,Suppressor ,Female ,Immunoglobulin Heavy Chains ,Developmental Biology ,Research Paper - Abstract
The INK4a/ARF locus encodes upstream regulators of the retinoblastoma and p53 tumor suppressor gene products. To compare the impact of these loci on tumor development and treatment response, the Emu-myc transgenic lymphoma model was used to generate genetically defined tumors with mutations in the INK4a/ARF, Rb, or p53 genes. Like p53 null lymphomas, INK4a/ARF null lymphomas formed rapidly, were highly invasive, displayed apoptotic defects, and were markedly resistant to chemotherapy in vitro and in vivo. Furthermore, INK4a/ARF(-/-) lymphomas displayed reduced p53 activity despite the presence of wild-type p53 genes. Consequently, INK4a/ARF and p53 mutations lead to aggressive tumors by disrupting overlapping tumor suppressor functions. These data have important implications for understanding the clinical behavior of human tumors.
- Published
- 1999
35. Characterization of novel potent and selective anaplastic lymphoma kinase (ALK) inhibitors
- Author
-
Chris Liang, E. de Stanchina, Hsuan-An Chen, William Pao, and Christine M. Lovly
- Subjects
Pathogenesis ,Cancer Research ,Oncology ,Crizotinib ,business.industry ,hemic and lymphatic diseases ,Cancer research ,Medicine ,Anaplastic lymphoma kinase ,business ,medicine.drug - Abstract
e13600 Background: Aberrant forms of the anaplastic lymphoma kinase (ALK) have been implicated in the pathogenesis of multiple human cancers and represent rational therapeutic targets. Crizotinib (...
- Published
- 2011
- Full Text
- View/download PDF
36. p53 action in apoptosis and senescence
- Author
-
Clemens A. Schmitt, Gerardo Ferbeyre, Scott W. Lowe, E. de Stanchina, Aw Lin, and M Soengas
- Subjects
Genome instability ,Senescence ,business.industry ,Cellular senescence ,Bioinformatics ,medicine.disease ,Breast cancer ,Action (philosophy) ,Apoptosis ,Surgical oncology ,Medicine ,Oral Presentation ,business ,Deleterious mutation - Published
- 2000
37. Concurrent SOS1 and MEK suppression inhibits signaling and growth of NF1-null melanoma.
- Author
-
Marasco M, Kumar D, Seale T, Borrego SG, Kaplun E, Aricescu I, Cole S, Qeriqi B, Qiu J, Chen X, Bahr A, Fidele D, Hofmann MH, Gerlach D, Savarese F, Merghoub T, Wolchok JD, Yao Z, de Stanchina E, Solit D, Misale S, and Rosen N
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Signal Transduction drug effects, Cell Proliferation drug effects, MAP Kinase Signaling System drug effects, Protein Kinase Inhibitors pharmacology, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Melanoma genetics, Melanoma pathology, Melanoma drug therapy, Melanoma metabolism, Neurofibromin 1 genetics, Neurofibromin 1 metabolism, SOS1 Protein metabolism, SOS1 Protein genetics
- Abstract
Neurofibromin (NF1) is a negative regulator of RAS signaling, frequently mutated in cancer. NF1-mutant melanoma is a highly malignant tumor for which targeted therapies are lacking. Here, we use biochemical and pharmacological assays on patient-derived models and isogenic cell lines to identify potential pharmacologic targets, revealing that NF1-null melanomas are dependent on RAS activation and that MEK inhibition relieves ERK-dependent negative feedback, increasing RAS signaling. MEK inhibition with avutometinib abrogates the adaptive rebound in ERK signaling, but the antitumor effects are limited. However, concurrent inhibition of MEK and SOS1 abrogates ERK activation, induces cell death, and suppresses tumor growth. In contrast to the NF1-deficient setting, concurrent SOS1 and SOS2 depletion is required to completely inhibit RAS signaling in NF1 wild-type cells. In sum, our data provide a mechanistic rationale for enhancing the therapeutic efficacy of MEK inhibitors by exploiting the lower residual SOS activity in NF1-null tumor cells., Competing Interests: Declaration of interests M.H.H., D.G., and F.S. are employees of Boehringer Ingelheim. T.M. is a consultant for Immunos Therapeutics, Daiichi Sankyo Co, TigaTX, Normunity, and Pfizer; is a cofounder of and equity holder in IMVAQ Therapeutics; has received research grants funding from Surface Oncology, Kyn Therapeutics, Infinity Pharmaceuticals, Peregrine Pharmaceuticals, Adaptive Biotechnologies, Leap Therapeutics, and Aprea Therapeutics; currently receives research funding from Bristol Myers Squibb, Enterome SA, and ReAlta Life Sciences; and is an inventor on patent applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neoantigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. J.D.W. is a consultant for Apricity, Ascentage Pharma, AstraZeneca, BeiGene, Bicara Therapeutics, Bristol Myers Squibb, Daiichi Sankyo, Dragonfly, Imvaq, Larkspur, Takeda, Tizona, Trishula Therapeutics, Immunocore (on their data safety board), and Scancell; received grant/research support from Bristol Myers Squibb and Enterome; has equity in Apricity, Arsenal IO/CellCarta, Ascentage, Imvaq, Linneaus, Larkspur, Georgiamune, Maverick, Tizona Therapeutics, and XenImmune; and is an inventor on the following patents: Xenogeneic DNA Vaccines, Newcastle Disease Viruses for Cancer Therapy, Myeloid-Derived Suppressor Cell Assay, Prediction of Responsiveness to Treatment with Immunomodulatory Therapeutics and Method of Monitoring Abscopal Effects During Such Treatment, Anti-PD1 Antibody, Anti-CTLA4 Antibodies, and Anti-GITR Antibodies and Methods of Use Thereof. Z.Y. is a past scientific advisory board (SAB) member of MapKure and currently an employee of Loxo Oncology at Lilly. D.S. reports personal fees from Pfizer, Vividion Therapeutics, BridgeBio, FogPharma, FORE Therapeutics, Scorpion Therapeutics, and Rain Therapeutics outside the submitted work. S.M. reports grants from Boehringer Ingelheim and Daiichi Sankyo outside the submitted work and is a SAB member of Bionseek. N.R. is on the SAB and owns equity in BeiGene, Zai Labs, MapKure, Ribon, and Effector; is also on the SAB of AstraZeneca and Chugai and a past SAB member of Novartis, Millennium-Takeda, Kura, and Araxes; and is a consultant to Revolution Medicines, Tarveda, Array-Pfizer, Boehringer Ingelheim, and Eli Lilly. He receives research funding from Revolution Medicines, AstraZeneca, Array Pfizer, and Boehringer Ingelheim and owns equity in Kura Oncology and Fortress., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
38. EGFR-directed antibodies promote HER2 ADC internalization and efficacy.
- Author
-
Gupta A, Michelini F, Shao H, Yeh C, Drago JZ, Liu D, Rosiek E, Romin Y, Ghafourian N, Thyparambil S, Misale S, Park W, de Stanchina E, Janjigian YY, Yaeger R, Li BT, and Chandarlapaty S
- Subjects
- Humans, Cell Line, Tumor, Animals, Mice, Immunoconjugates pharmacology, Female, Xenograft Model Antitumor Assays, Mice, Nude, ErbB Receptors metabolism, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Endocytosis drug effects, Trastuzumab pharmacology
- Abstract
Trastuzumab deruxtecan (T-DXd) is a human epidermal growth factor receptor 2 (HER2)-targeting antibody drug conjugate that has remarkable activity in HER2-positive cancers. However, the degree of benefit of T-DXd is not uniform among solid tumors even with high levels of HER2. Despite high HER2 expression, the HER2/T-DXd complex may not always undergo internalization and payload release dependent on the receptor's conformation and context. We hypothesize that epidermal growth factor receptor (EGFR), a dimerization partner of HER2, can modulate HER2 trafficking through endocytic pathways and affect T-DXd uptake. We demonstrate that elevated EGFR expression levels can promote EGFR/HER2 heterodimer formation and suppress T-DXd internalization and efficacy. Knockdown of EGFR expression or pharmacologic stimulation of EGFR endocytosis with EGFR monoclonal antibodies restores T-DXd trafficking and antitumor activity in EGFR-overexpressing cancers in vivo. Our results reveal EGFR overexpression to be a potential mechanism of resistance to T-DXd, which can be overcome by combination therapy strategies targeting EGFR., Competing Interests: Declaration of interests F.M. is employed by AstraZeneca and reports stock and other ownership interests from AstraZeneca. J.Z.D. reports research funding from AstraZeneca and consulting fees from AstraZeneca, Genagon, AmMax Bio, NuProbe, and Launchpad Therapeutics. S.T. and N.G. are employed by mProbe and report research funding, stock, and other ownership interests from mProbe. S.M. reports grants from Boehringer Ingelheim and Daiichi Sankyo and serves on the scientific advisory board of Bionseek. Y.Y.J. acknowledges research funding from the National Cancer Institute, Department of Defense, Cycle for Survival, Fred’s Team, Inspirna, AstraZeneca, Arcus Biosciences, Bayer, Genentech/Roche, Bristol-Myers Squibb, Eli Lilly, Merck, and Transcenta. She has served on advisory boards and/or done consulting for AbbVie, AmerisourceBergen, AskGene Pharma, Inc., Arcus Biosciences, Astellas, AstraZeneca, Basilea Pharmaceutica, Bayer, Bristol-Myers Squibb, Daiichi Sanyko, Eli Lilly, Geneos Therapeutics, GlaxoSmithKline, Guardant Health, Inc., Imedex, Imugene, Inspirna, Lynx Health, Merck, Merck Serono, Mersana Therapeutics, Pfizer, Seagen, Silverback Therapeutics, and Zymeworks, Inc., as well as Clinical Care Options, HMP Education, Michael J. Hennessy Associates, Paradigm Medical Communications, PeerView Institute, and Research to Practice. She also holds stock options in Inspirna. W.P. acknowledges research funding from the National Cancer Institute, Break Through Cancer, Parker Institute for Cancer Immunotherapy (PICI), Society for Immunotherapy of Cancer, The Society of MSK, Merck, Astellas, Miracogen, Amgen, and Revolution Medicines. He has served on advisory boards and/or done consulting for Astellas, EXACT Therapeutics, and Innovent Biologics. He has received honoraria for continuing medical education for American Physician Institute and Integrity. R.Y. has served as an advisor for Array BioPharma/Pfizer, Mirati Therapeutics, and Amgen; received a speaker’s honorarium from Zai Lab; and has received research support to her institution from Array BioPharma/Pfizer, Boehringer Ingelheim, Boundless Bio, Mirati Therapeutics, and Daiichi Sankyo. B.T.L. reports grants and other support from Amgen and grants from the NIH during the conduct of the study; grants and personal fees from Hengrui USA; grants and other support from Genentech Roche, Lilly, AstraZeneca, and Daiichi Sankyo; nonfinancial support and other support from Resolution Bioscience; grants and nonfinancial support from MORE Health; nonfinancial support from Jiangsu Hengrui Medicine; personal fees from Boehringer Ingelheim; grants from the NIH outside the submitted work; and a patent for US62/685,057 issued, a patent for US62/514,661 issued, royalties from Karger Publishers, and licensing with Shanghai Jiao Tong University Press. S.C. has received personal consulting fees from Novartis, AstraZeneca, Nuvalent, Genesis Therapeutics, eFFECTOR Therapeutics, SAGA Diagnostics, Prelude Therapeutics, and Casdin Capital. S.C. has research funding from AstraZeneca and Daiichi Sankyo., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
39. uPAR Immuno-PET in Pancreatic Cancer, Aging, and Chemotherapy-Induced Senescence.
- Author
-
Pratt EC, Mezzadra R, Kulick A, Kaminsky S, Samuels ZV, Loor A, de Stanchina E, Lowe SW, and Lewis JS
- Subjects
- Animals, Mice, Humans, Cell Line, Tumor, Aging, Pyridines pharmacology, Cellular Senescence drug effects, Piperazines pharmacology, Piperazines therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms drug therapy, Receptors, Urokinase Plasminogen Activator metabolism, Positron-Emission Tomography
- Abstract
Identifying cancer therapy resistance is a key time-saving tool for physicians. Part of chemotherapy resistance includes senescence, a persistent state without cell division or cell death. Chemically inducing senescence with the combination of trametinib and palbociclib (TP) yields several tumorigenic and prometastatic factors in pancreatic cancer models with many potential antibody-based targets. In particular, urokinase plasminogen activator receptor (uPAR) has been shown to be a membrane-bound marker of senescence in addition to an oncology target. Methods: Here, 2 antibodies against murine uPAR and human uPAR were developed as immuno-PET agents to noninvasively track uPAR antigen abundance. Results: TP treatment increased cell uptake both in murine KPC cells and in human MiaPaCa2 cells. In vivo, subcutaneously implanted murine KPC tumors had high tumor uptake with the antimurine uPAR antibody independently of TP in young mice, yet uPAR uptake was maintained in aged mice on TP. Mice xenografted with human MiaPaCa2 tumors showed a significant increase in tumor uptake on TP therapy when imaged with the antihuman uPAR antibody. Imaging with either uPAR antibody was found to be more tumor-selective than imaging with [
18 F]FDG or [18 F]F-DPA-714. Conclusion: The use of radiolabeled uPAR-targeting antibodies provides a new antibody-based PET imaging candidate for pancreatic cancer imaging as well as chemotherapy-induced senescence., (© 2024 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2024
- Full Text
- View/download PDF
40. A Classical Epithelial State Drives Acute Resistance to KRAS Inhibition in Pancreatic Cancer.
- Author
-
Singhal A, Styers HC, Rub J, Li Z, Torborg SR, Kim JY, Grbovic-Huezo O, Feng H, Tarcan ZC, Sahin Ozkan H, Hallin J, Basturk O, Yaeger R, Christensen JG, Betel D, Yan Y, Chio IIC, de Stanchina E, and Tammela T
- Subjects
- Mice, Humans, Animals, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Xenograft Model Antitumor Assays, Proto-Oncogene Proteins p21(ras) genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Drug Resistance, Neoplasm
- Abstract
Intratumoral heterogeneity in pancreatic ductal adenocarcinoma (PDAC) is characterized by a balance between basal and classical epithelial cancer cell states, with basal dominance associating with chemoresistance and a dismal prognosis. Targeting oncogenic KRAS, the primary driver of pancreatic cancer, shows early promise in clinical trials, but efficacy is limited by acquired resistance. Using genetically engineered mouse models and patient-derived xenografts, we find that basal PDAC cells are highly sensitive to KRAS inhibitors. Employing fluorescent and bioluminescent reporter systems, we longitudinally track cell-state dynamics in vivo and reveal a rapid, KRAS inhibitor-induced enrichment of the classical state. Lineage tracing uncovers that these enriched classical PDAC cells are a reservoir for disease relapse. Genetic or chemotherapy-mediated ablation of the classical cell state is synergistic with KRAS inhibition, providing a preclinical proof of concept for this therapeutic strategy. Our findings motivate combining classical state-directed therapies with KRAS inhibitors to deepen responses and counteract resistance in pancreatic cancer. Significance: KRAS inhibitors hold promise in pancreatic cancer, but responses are limited by acquired resistance. We find that a classical epithelial cancer cell state is acutely resistant to KRAS inhibition and serves as a reservoir for disease relapse. Targeting the classical state alongside KRAS inhibition deepens responses, revealing a potent therapeutic strategy. See related commentary by Marasco and Misale, p. 2018., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
41. Pharmacologic restoration of GTP hydrolysis by mutant RAS.
- Author
-
Cuevas-Navarro A, Pourfarjam Y, Hu F, Rodriguez DJ, Vides A, Sang B, Fan S, Goldgur Y, de Stanchina E, and Lito P
- Abstract
Approximately 3.4 million patients worldwide are diagnosed each year with cancers that harbor pathogenic mutations in one of three RAS proto-oncogenes (KRAS, NRAS and HRAS)
1,2 . These mutations impair the GTPase activity of RAS, leading to activation of downstream signaling and proliferation3-6 . Long-standing efforts to restore the hydrolase activity of RAS mutants have been unsuccessful, extinguishing any consideration towards a viable therapeutic strategy7 . Here we show that tri-complex inhibitors, that is, molecular glues with the ability to recruit cyclophilin A (CYPA) to the active state of RAS have a dual mechanism of action: not only do they prevent activated RAS from binding to its effectors, but, unexpectedly, they also stimulate GTP hydrolysis. Drug-bound CYPA complexes modulate residues in the switch II motif of RAS to coordinate the nucleophilic attack on the ɣ phosphate of GTP, in a mutation-specific manner. RAS mutants most sensitive to stimulation of GTPase activity were more susceptible to treatment compared to mutants whose hydrolysis could not be enhanced, suggesting that pharmacologic stimulation of hydrolysis potentiates the therapeutic effects of tri-complex inhibitors for specific RAS mutants. This study lays the foundation for developing a new class of therapeutics that inhibit cancer growth by stimulating mutant GTPase activity., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
42. Phosphorylation-driven epichaperome assembly is a regulator of cellular adaptability and proliferation.
- Author
-
Roychowdhury T, McNutt SW, Pasala C, Nguyen HT, Thornton DT, Sharma S, Botticelli L, Digwal CS, Joshi S, Yang N, Panchal P, Chakrabarty S, Bay S, Markov V, Kwong C, Lisanti J, Chung SY, Ginsberg SD, Yan P, De Stanchina E, Corben A, Modi S, Alpaugh ML, Colombo G, Erdjument-Bromage H, Neubert TA, Chalkley RJ, Baker PR, Burlingame AL, Rodina A, Chiosis G, and Chu F
- Subjects
- Humans, Phosphorylation, Animals, Protein Interaction Maps, Mice, Serine metabolism, Cell Line, Tumor, Cell Proliferation, HSP90 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins genetics, Molecular Chaperones metabolism, Molecular Chaperones genetics, Protein Processing, Post-Translational
- Abstract
The intricate network of protein-chaperone interactions is crucial for maintaining cellular function. Recent discoveries have unveiled the existence of specialized chaperone assemblies, known as epichaperomes, which serve as scaffolding platforms that orchestrate the reconfiguration of protein-protein interaction networks, thereby enhancing cellular adaptability and proliferation. This study explores the structural and regulatory aspects of epichaperomes, with a particular focus on the role of post-translational modifications (PTMs) in their formation and function. A key finding is the identification of specific PTMs on HSP90, particularly at residues Ser226 and Ser255 within an intrinsically disordered region, as critical determinants of epichaperome assembly. Our data demonstrate that phosphorylation of these serine residues enhances HSP90's interactions with other chaperones and co-chaperones, creating a microenvironment conducive to epichaperome formation. Moreover, we establish a direct link between epichaperome function and cellular physiology, particularly in contexts where robust proliferation and adaptive behavior are essential, such as in cancer and pluripotent stem cell maintenance. These findings not only provide mechanistic insights but also hold promise for the development of novel therapeutic strategies targeting chaperone assemblies in diseases characterized by epichaperome dysregulation, thereby bridging the gap between fundamental research and precision medicine., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
43. HER2 Antibody-Drug Conjugates Are Active against Desmoplastic Small Round Cell Tumor.
- Author
-
Zhang T, Febres-Aldana CA, Liu Z, Dix JM, Cheng R, Dematteo RG, Lui AJW, Khodos I, Gili L, Mattar MS, Lisanti J, Kwong C, Linkov I, Tipping MJ, de Stanchina E, Odintsov I, Ladanyi M, and Somwar R
- Subjects
- Humans, Animals, Mice, Male, Cell Line, Tumor, Ado-Trastuzumab Emtansine pharmacology, Ado-Trastuzumab Emtansine therapeutic use, Female, Apoptosis drug effects, Camptothecin analogs & derivatives, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 genetics, Desmoplastic Small Round Cell Tumor drug therapy, Desmoplastic Small Round Cell Tumor pathology, Desmoplastic Small Round Cell Tumor genetics, Desmoplastic Small Round Cell Tumor metabolism, Immunoconjugates pharmacology, Immunoconjugates therapeutic use, Xenograft Model Antitumor Assays, Trastuzumab pharmacology, Trastuzumab therapeutic use
- Abstract
Purpose: Desmoplastic small round cell tumor (DSRCT) is a rare but highly aggressive soft tissue sarcoma that arises in the abdominopelvic cavity of young males. Since the discovery of EWSR1::WT1 fusion as the driver of DSRCT, no actionable genomic alterations have been identified, limiting disease management to a combination of surgery, chemotherapy, and radiation, with very poor outcomes. Herein, we evaluated ERBB2/HER2 expression in DSRCT as a therapeutic target., Experimental Design: ERBB2/HER2 expression was assessed in clinical samples and patient-derived xenografts (PDX) using RNA sequencing, RT-qPCR, and a newly developed HER2 IHC assay (clone 29D8). Responses to HER2 antibody-drug conjugates (ADC)-trastuzumab deruxtecan (T-DXd) and trastuzumab emtansine-were evaluated in DSRCT PDX, cell line, and organoid models. Drug internalization was demonstrated by live microscopy. Apoptosis was evaluated by Western blotting and caspase activity assays., Results: ERBB2/HER2 was detectable in DSRCT samples from patients and PDXs, with higher sensitivity RNA assays and improved IHC detectability using clone 29D8. Treatment of ERBB2/HER2-expressing DSRCT PDX, cell line, and organoid models with T-DXd or trastuzumab emtansine resulted in tumor regression. This therapeutic response was long-lasting in T-DXd-treated xenografts and was mediated by rapid HER2 ADC complex internalization and cytotoxicity, triggering p53-mediated apoptosis and growth arrest. Xenograft regression was associated with bystander payload effects triggering global tumor niche responses proportional to HER2 status., Conclusions: ERBB2/HER2 is a therapeutic target in DSRCT. HER2 ADCs may represent novel options for managing this exceptionally aggressive sarcoma, possibly fulfilling an urgent and historically unmet need for more effective clinical therapy., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
44. Targetable treatment resistance in thyroid cancer with clonal hematopoiesis.
- Author
-
Tiedje V, Vela PS, Yang JL, Untch BR, Boucai L, Stonestrom AJ, Costa AB, Expósito SF, Srivastava A, Kerpelev M, Greenberg J, Wereski M, Kulick A, Chen K, Qin T, Im SY, Krishnan A, Martinez Benitez AR, Pluvinet R, Sahin M, Menghrajani K, Krishnamoorthy GP, de Stanchina E, Zehir A, Satija R, Knauf J, Bowman RL, Esteller M, Devlin S, Berger MF, Koche RP, Fagin JA, and Levine RL
- Abstract
Anaplastic thyroid cancer (ATC) is a clinically aggressive malignancy with a dismal prognosis. Combined BRAF/MEK inhibition offers significant therapeutic benefit in patients with BRAF
V600E -mutant ATCs. However, relapses are common and overall survival remains poor. Compared with differentiated thyroid cancer, a hallmark of ATCs is significant infiltration with myeloid cells, particularly macrophages. ATCs are most common in the aging population, which also has an increased incidence of TET2 -mutant clonal hematopoiesis (CH). CH-mutant macrophages have been shown to accelerate CH-associated pathophysiology including atherosclerosis. However, the clinical and mechanistic contribution of CH-mutant clones to solid tumour biology, prognosis and therapeutic response has not been elucidated. Here we show that TET2 -mutant CH is enriched in the tumour microenvironment of patients with solid tumours and associated with adverse prognosis in ATC patients. We find that Tet2 -mutant macrophages selectively infiltrate mouse BrafV600E -mutant ATC and that their overexpression of Tgfβ-family ligands mediates resistance to BRAF/MEK inhibition. Importantly, inhibition of Tgfβ signaling restores sensitivity to MAPK pathway inhibition, opening a path for synergistic strategies to improve outcomes of patients with ATCs and concurrent CH.- Published
- 2024
- Full Text
- View/download PDF
45. SETD1B mutations confer apoptosis resistance and BCL2 independence in B cell lymphoma.
- Author
-
Portelinha A, Wang S, Parsa S, Jiang M, Gorelick AN, Mohanty S, Sharma S, de Stanchina E, Berishaj M, Zhao C, Heward J, Aryal NK, Tavana O, Wen J, Fitzgibbon J, Dogan A, Younes A, Melnick AM, and Wendel HG
- Subjects
- Animals, Humans, Mice, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Line, Tumor, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Drug Resistance, Neoplasm genetics, Lymphoma, B-Cell genetics, Lymphoma, B-Cell pathology, Lymphoma, B-Cell metabolism, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Lymphoma, Large B-Cell, Diffuse metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Sulfonamides pharmacology, Apoptosis genetics, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Mutation, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
The translocation t(14;18) activates BCL2 and is considered the initiating genetic lesion in most follicular lymphomas (FL). Surprisingly, FL patients fail to respond to the BCL2 inhibitor, Venetoclax. We show that mutations and deletions affecting the histone lysine methyltransferase SETD1B (KMT2G) occur in 7% of FLs and 16% of diffuse large B cell lymphomas (DLBCL). Deficiency in SETD1B confers striking resistance to Venetoclax and an experimental MCL-1 inhibitor. SETD1B also acts as a tumor suppressor and cooperates with the loss of KMT2D in lymphoma development in vivo. Consistently, loss of SETD1B in human lymphomas typically coincides with loss of KMT2D. Mechanistically, SETD1B is required for the expression of several proapoptotic BCL2 family proteins. Conversely, inhibitors of the KDM5 histone H3K4 demethylases restore BIM and BIK expression and synergize with Venetoclax in SETD1B-deficient lymphomas. These results establish SETD1B as an epigenetic regulator of cell death and reveal a pharmacological strategy to augment Venetoclax sensitivity in lymphoma., (© 2024 Portelinha et al.)
- Published
- 2024
- Full Text
- View/download PDF
46. Augmenting CAR T-cell Functions with LIGHT.
- Author
-
Cai W, Tanaka K, Mi X, Rajasekhar VK, Khan JF, Yoo S, de Stanchina E, Rahman J, Mathew S, Abrahimi P, Souness S, Purdon TJ, McDowell JR, Meyerberg J, Fujino T, Healey JH, Abdel-Wahab O, Scheinberg DA, Brentjens RJ, and Daniyan AF
- Subjects
- Humans, Animals, Cell Line, Tumor, Cytotoxicity, Immunologic, Mice, Lymphotoxin beta Receptor immunology, Lymphotoxin beta Receptor metabolism, Antigens, Neoplasm immunology, Xenograft Model Antitumor Assays, Neoplasms immunology, Neoplasms therapy, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Immunotherapy, Adoptive methods, Tumor Necrosis Factor Ligand Superfamily Member 14 metabolism, T-Lymphocytes immunology
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy has resulted in remarkable clinical success in the treatment of B-cell malignancies. However, its clinical efficacy in solid tumors is limited, primarily by target antigen heterogeneity. To overcome antigen heterogeneity, we developed CAR T cells that overexpress LIGHT, a ligand of both lymphotoxin-β receptor on cancer cells and herpes virus entry mediator on immune cells. LIGHT-expressing CAR T cells displayed both antigen-directed cytotoxicity mediated by the CAR and antigen-independent killing mediated through the interaction of LIGHT with lymphotoxin-β receptor on cancer cells. Moreover, CAR T cells expressing LIGHT had immunostimulatory properties that improved the cells' proliferation and cytolytic profile. These data indicate that LIGHT-expressing CAR T cells may provide a way to eliminate antigen-negative tumor cells to prevent antigen-negative disease relapse., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
47. Correction: SMARCA4 controls state plasticity in small cell lung cancer through regulation of neuroendocrine transcription factors and REST splicing.
- Author
-
Redin E, Sridhar H, Zhan YA, Pereira Mello B, Zhong H, Durani V, Sabet A, Manoj P, Linkov I, Qiu J, Koche RP, de Stanchina E, Astorkia M, Betel D, Quintanal-Villalonga Á, and Rudin CM
- Published
- 2024
- Full Text
- View/download PDF
48. ATR inhibition activates cancer cell cGAS/STING-interferon signaling and promotes antitumor immunity in small-cell lung cancer.
- Author
-
Taniguchi H, Chakraborty S, Takahashi N, Banerjee A, Caeser R, Zhan YA, Tischfield SE, Chow A, Nguyen EM, Villalonga ÁQ, Manoj P, Shah NS, Rosario S, Hayatt O, Qu R, de Stanchina E, Chan J, Mukae H, Thomas A, Rudin CM, and Sen T
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Interferons metabolism, B7-H1 Antigen metabolism, B7-H1 Antigen antagonists & inhibitors, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Topotecan pharmacology, Pyrazines pharmacology, Pyrazines therapeutic use, Isoxazoles, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Ataxia Telangiectasia Mutated Proteins metabolism, Nucleotidyltransferases metabolism, Lung Neoplasms drug therapy, Lung Neoplasms immunology, Lung Neoplasms metabolism, Lung Neoplasms pathology, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma immunology, Small Cell Lung Carcinoma metabolism, Small Cell Lung Carcinoma pathology, Membrane Proteins metabolism, Membrane Proteins genetics, Signal Transduction drug effects
- Abstract
Patients with small-cell lung cancer (SCLC) have poor prognosis and typically experience only transient benefits from combined immune checkpoint blockade (ICB) and chemotherapy. Here, we show that inhibition of ataxia telangiectasia and rad3 related (ATR), the primary replication stress response activator, induces DNA damage-mediated micronuclei formation in SCLC models. ATR inhibition in SCLC activates the stimulator of interferon genes (STING)-mediated interferon signaling, recruits T cells, and augments the antitumor immune response of programmed death-ligand 1 (PD-L1) blockade in mouse models. We demonstrate that combined ATR and PD-L1 inhibition causes improved antitumor response than PD-L1 alone as the second-line treatment in SCLC. This study shows that targeting ATR up-regulates major histocompatibility class I expression in preclinical models and SCLC clinical samples collected from a first-in-class clinical trial of ATR inhibitor, berzosertib, with topotecan in patients with relapsed SCLC. Targeting ATR represents a transformative vulnerability of SCLC and is a complementary strategy to induce STING-interferon signaling-mediated immunogenicity in SCLC.
- Published
- 2024
- Full Text
- View/download PDF
49. Combination of MDM2 and Targeted Kinase Inhibitors Results in Prolonged Tumor Control in Lung Adenocarcinomas With Oncogenic Tyrosine Kinase Drivers and MDM2 Amplification.
- Author
-
Elkrief A, Odintsov I, Smith RS, Vojnic M, Hayashi T, Khodos I, Markov V, Liu Z, Lui AJW, Bloom JL, Offin MD, Rudin CM, de Stanchina E, Riely GJ, Somwar R, and Ladanyi M
- Subjects
- Humans, Animals, Mice, Gene Amplification, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Adenocarcinoma of Lung drug therapy, Adenocarcinoma of Lung genetics, Protein Kinase Inhibitors therapeutic use
- Abstract
Purpose: MDM2, a negative regulator of the TP53 tumor suppressor, is oncogenic when amplified. MDM2 amplification (MDM2amp) is mutually exclusive with TP53 mutation and is seen in 6% of patients with lung adenocarcinoma (LUAD), with significant enrichment in subsets with receptor tyrosine kinase (RTK) driver alterations. Recent studies have shown synergistic activity of MDM2 and MEK inhibition in patient-derived LUAD models with MDM2amp and RTK driver alterations. However, the combination of MDM2 and RTK inhibitors in LUAD has not been studied., Methods: We evaluated the combination of MDM2 and RTK inhibition in patient-derived models of LUAD., Results: In a RET-fusion LUAD patient-derived model with MDM2amp, MDM2 inhibition with either milademetan or AMG232 combined with selpercatinib resulted in long-term in vivo tumor control markedly superior to either agent alone. Similarly, in an EGFR-mutated model with MDM2amp, combining either milademetan or AMG232 with osimertinib resulted in long-term in vivo tumor control, which was strikingly superior to either agent alone., Conclusion: These preclinical in vivo data provide a rationale for further clinical development of this combinatorial targeted therapy approach.
- Published
- 2024
- Full Text
- View/download PDF
50. TYRP1 directed CAR T cells control tumor progression in preclinical melanoma models.
- Author
-
Hackett CS, Hirschhorn D, Tang MS, Purdon TJ, Marouf Y, Piersigilli A, Agaram NP, Liu C, Schad SE, de Stanchina E, Rafiq S, Monette S, Wolchok JD, Merghoub T, and Brentjens RJ
- Abstract
Despite therapeutic efficacy observed with immune checkpoint blockade in advanced melanoma, many tumors do not respond to treatment, representing a need for new therapies. Here, we have generated chimeric antigen receptor (CAR) T cells targeting TYRP1, a melanoma differentiation antigen expressed on the surface of melanomas, including rare acral and uveal melanomas. TYRP1-targeted CAR T cells demonstrate antigen-specific activation and cytotoxic activity in vitro and in vivo against human melanomas independent of the MHC alleles and expression. In addition, the toxicity to pigmented normal tissues observed with T lymphocytes expressing TYRP1-targeted TCRs was not observed with TYRP1-targeted CAR T cells. Anti-TYRP1 CAR T cells provide a novel means to target advanced melanomas, serving as a platform for the development of similar novel therapeutic agents and as a tool to interrogate the immunobiology of melanomas., Competing Interests: C.S.H., D.H., T.J.P., S.R., J.D.W., T.M., and R.J.B. are inventors on multiple patents filed by MSK covering CAR T cell technology, including the CAR T cells discussed in this manuscript. S.R. serves on the Scientific Advisory Board of Celyad Oncology. R.J.B. has licensed intellectual property to and collects royalties from BMS, Caribou, and Sanofi. R.J.B. received research funding from BMS. R.J.B. is a consultant to BMS, Atara Biotherapeutics Inc., and Triumvira, and was a consultant for Cargo Tx and CoImmune but ended in the past 3 months, and Gracell Biotechnologies Inc. but ended employment in the past 24 months. R.J.B. is a member of the scientific advisory board for Triumvira and was a member of the scientific advisory board for Cargo Tx and CoImmune, but that ended in the past 6 months. J.D.W. is a consultant for Apricity, Ascentage Pharma, AstraZeneca, BeiGene, Bicara Therapeutics (ending 4/1/2024), Bristol Myers Squibb, Daiichi Sankyo, Dragonfly, Imvaq, Larkspur, Psioxus, Recepta, Takeda, Tizona, Trishula Therapeutics, and Sellas. J.D.W. received grant/research support from Bristol Myers Squibb and Enterome. J.D.W. has equity in Apricity, Arsenal IO/CellCarta, Ascentage, Imvaq, Linneaus, Larkspur, Georgiamune, Maverick, Tizona Therapeutics, and Xenimmune. J.D.W. is an inventor on the following patents: Xenogeneic DNA Vaccines, Newcastle Disease viruses for Cancer Therapy, Myeloid-derived suppressor cell (MDSC) assay, Prediction of responsiveness to treatment with immunomodulatory therapeutics and method of monitoring abscopal effects during such treatment, Anti-PD1 Antibody, Anti-CTLA4 antibodies, Anti-GITR antibodies and methods of use thereof. T.M. is a consultant for Immunos Therapeutics, Daiichi Sankyo Co, TigaTX, Normunity, and Pfizer. T.M. is a cofounder of and equity holder in Imvaq Therapeutics. T.M. receives research grant funding from Bristol Myers Squibb, Surface Oncology, Kyn Therapeutics, Infinity Pharmaceuticals, Peregrine Pharmaceuticals, Adaptive Biotechnologies, Leap Therapeutics, and Aprea Therapeutics. T.M. is an inventor on patent applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neo-antigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4., (© 2024 The Author(s). Published by Elsevier Inc. on behalf of The American Society of Gene and Cell Therapy.)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.