28 results on '"Rebecca Caeser"'
Search Results
2. WEE1 inhibition enhances the antitumor immune response to PD-L1 blockade by the concomitant activation of STING and STAT1 pathways in SCLC
- Author
-
Hirokazu Taniguchi, Rebecca Caeser, Shweta S. Chavan, Yingqian A. Zhan, Andrew Chow, Parvathy Manoj, Fathema Uddin, Hidenori Kitai, Rui Qu, Omar Hayatt, Nisargbhai S. Shah, Álvaro Quintanal Villalonga, Viola Allaj, Evelyn M. Nguyen, Joseph Chan, Adam O. Michel, Hiroshi Mukae, Elisa de Stanchina, Charles M. Rudin, and Triparna Sen
- Subjects
CP: Cancer ,CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Small cell lung cancers (SCLCs) have high mutational burden but are relatively unresponsive to immune checkpoint blockade (ICB). Using SCLC models, we demonstrate that inhibition of WEE1, a G2/M checkpoint regulator induced by DNA damage, activates the STING-TBK1-IRF3 pathway, which increases type I interferons (IFN-α and IFN-β) and pro-inflammatory chemokines (CXCL10 and CCL5), facilitating an immune response via CD8+ cytotoxic T cell infiltration. We further show that WEE1 inhibition concomitantly activates the STAT1 pathway, increasing IFN-γ and PD-L1 expression. Consistent with these findings, combined WEE1 inhibition (AZD1775) and PD-L1 blockade causes remarkable tumor regression, activation of type I and II interferon pathways, and infiltration of cytotoxic T cells in multiple immunocompetent SCLC genetically engineered mouse models, including an aggressive model with stabilized MYC. Our study demonstrates cell-autonomous and immune-stimulating activity of WEE1 inhibition in SCLC models. Combined inhibition of WEE1 plus PD-L1 blockade represents a promising immunotherapeutic approach in SCLC.
- Published
- 2022
- Full Text
- View/download PDF
3. MAPK pathway activation selectively inhibits ASCL1-driven small cell lung cancer
- Author
-
Rebecca Caeser, Christopher Hulton, Emily Costa, Vidushi Durani, Megan Little, Xiaoping Chen, Sam E. Tischfield, Marina Asher, Faruk Erdem Kombak, Shweta S. Chavan, Nisargbhai S. Shah, Metamia Ciampricotti, Elisa de Stanchina, John T. Poirier, Charles M. Rudin, and Triparna Sen
- Subjects
Biological sciences ,Cell biology ,Molecular biology ,Science - Abstract
Summary: Activation of mitogenic signaling pathways is a common oncogenic driver of many solid tumors including lung cancer. Although activating mutations in the mitogen-activated protein kinase (MAPK) pathway are prevalent in non-small cell lung cancers, MAPK pathway activity, counterintuitively, is relatively suppressed in the more aggressively proliferative small cell lung cancer (SCLC). Here, we elucidate the role of the MAPK pathway and how it interacts with other signaling pathways in SCLC. We find that the most common SCLC subtype, SCLC-A associated with high expression of ASCL1, is selectively sensitive to MAPK activation in vitro and in vivo through induction of cell-cycle arrest and senescence. We show strong upregulation of ERK negative feedback regulators and STAT signaling upon MAPK activation in SCLC-A lines. These findings provide insight into the complexity of signaling networks in SCLC and suggest subtype-specific mitogenic vulnerabilities.
- Published
- 2021
- Full Text
- View/download PDF
4. Genetic modification of primary human B cells to model high-grade lymphoma
- Author
-
Rebecca Caeser, Miriam Di Re, Joanna A. Krupka, Jie Gao, Maribel Lara-Chica, João M. L. Dias, Susanna L. Cooke, Rachel Fenner, Zelvera Usheva, Hendrik F. P. Runge, Philip A. Beer, Hesham Eldaly, Hyo-Kyung Pak, Chan-Sik Park, George S. Vassiliou, Brian J. P. Huntly, Annalisa Mupo, Rachael J. M. Bashford-Rogers, and Daniel J. Hodson
- Subjects
Science - Abstract
A dearth of adequate preclinical models to faithfully mimic diffuse large B-cell lymphoma has hampered the identification of driver genes. Here, the authors present a co-culture system that enables ex vivo expansion, viral transduction and transformation of primary human germinal center B cells.
- Published
- 2019
- Full Text
- View/download PDF
5. cfDNA methylome profiling for detection and subtyping of small cell lung cancers
- Author
-
Francesca Chemi, Simon P. Pearce, Alexandra Clipson, Steven M. Hill, Alicia-Marie Conway, Sophie A. Richardson, Katarzyna Kamieniecka, Rebecca Caeser, Daniel J. White, Sumitra Mohan, Victoria Foy, Kathryn L. Simpson, Melanie Galvin, Kristopher K. Frese, Lynsey Priest, Jacklynn Egger, Alastair Kerr, Pierre P. Massion, John T. Poirier, Gerard Brady, Fiona Blackhall, Dominic G. Rothwell, Charles M. Rudin, and Caroline Dive
- Subjects
Mice ,Epigenome ,Cancer Research ,Lung Neoplasms ,Oncology ,Animals ,DNA Methylation ,Cell-Free Nucleic Acids ,Small Cell Lung Carcinoma ,Transcription Factors - Abstract
Small cell lung cancer (SCLC) is characterized by morphologic, epigenetic and transcriptomic heterogeneity. Subtypes based upon predominant transcription factor expression have been defined that, in mouse models and cell lines, exhibit potential differential therapeutic vulnerabilities, with epigenetically distinct SCLC subtypes also described. The clinical relevance of these subtypes is unclear, due in part to challenges in obtaining tumor biopsies for reliable profiling. Here we describe a robust workflow for genome-wide DNA methylation profiling applied to both patient-derived models and to patients’ circulating cell-free DNA (cfDNA). Tumor-specific methylation patterns were readily detected in cfDNA samples from patients with SCLC and were correlated with survival outcomes. cfDNA methylation also discriminated between the transcription factor SCLC subtypes, a precedent for a liquid biopsy cfDNA-methylation approach to molecularly subtype SCLC. Our data reveal the potential clinical utility of cfDNA methylation profiling as a universally applicable liquid biopsy approach for the sensitive detection, monitoring and molecular subtyping of patients with SCLC.
- Published
- 2022
6. Combination therapy with MDM2 and MEK inhibitors is effective in patient-derived models of lung adenocarcinoma with concurrent oncogenic drivers and MDM2 amplification
- Author
-
Arielle Elkrief, Igor Odintsov, Vladimir Markov, Rebecca Caeser, Pawel Sobczuk, Sam E. Tischfield, Umesh Bhanot, Chad M. Vanderbilt, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Vijaya G. Tirunagaru, Robert C. Doebele, Álvaro Quintanal-Villalonga, Charles M. Rudin, Romel Somwar, and Marc Ladanyi
- Subjects
Pulmonary and Respiratory Medicine ,Oncology - Published
- 2023
7. Supplementary Figures S1 - S5 from Rlf–Mycl Gene Fusion Drives Tumorigenesis and Metastasis in a Mouse Model of Small Cell Lung Cancer
- Author
-
Charles M. Rudin, Thales Papagiannakopoulos, John T. Poirier, Trudy G. Oliver, Triparna Sen, Mark T.A. Donoghue, Tyler Jacks, Elliot H. Akama-Garren, William M. Rideout, Andrea Ventura, Danilo Maddalo, Anastasia-Maria Zavitsanou, Janneke E. Jaspers, Francisco J. Sanchez-Rivera, Faruk E. Kombak, Kyle B. Spainhower, Parvathy Manoj, Viola Allaj, Emily A. Costa, Rebecca Caeser, Angeliki Karatza, Àlvaro Quintanal-Villalonga, Allison L. Richards, Triantafyllia Karakousi, and Metamia Ciampricotti
- Abstract
Supplementary Figures S1 - S5
- Published
- 2023
8. Data from Rlf–Mycl Gene Fusion Drives Tumorigenesis and Metastasis in a Mouse Model of Small Cell Lung Cancer
- Author
-
Charles M. Rudin, Thales Papagiannakopoulos, John T. Poirier, Trudy G. Oliver, Triparna Sen, Mark T.A. Donoghue, Tyler Jacks, Elliot H. Akama-Garren, William M. Rideout, Andrea Ventura, Danilo Maddalo, Anastasia-Maria Zavitsanou, Janneke E. Jaspers, Francisco J. Sanchez-Rivera, Faruk E. Kombak, Kyle B. Spainhower, Parvathy Manoj, Viola Allaj, Emily A. Costa, Rebecca Caeser, Angeliki Karatza, Àlvaro Quintanal-Villalonga, Allison L. Richards, Triantafyllia Karakousi, and Metamia Ciampricotti
- Abstract
Small cell lung cancer (SCLC) has limited therapeutic options and an exceptionally poor prognosis. Understanding the oncogenic drivers of SCLC may help define novel therapeutic targets. Recurrent genomic rearrangements have been identified in SCLC, most notably an in-frame gene fusion between RLF and MYCL found in up to 7% of the predominant ASCL1-expressing subtype. To explore the role of this fusion in oncogenesis and tumor progression, we used CRISPR/Cas9 somatic editing to generate a Rlf–Mycl-driven mouse model of SCLC. RLF–MYCL fusion accelerated transformation and proliferation of murine SCLC and increased metastatic dissemination and the diversity of metastatic sites. Tumors from the RLF–MYCL genetically engineered mouse model displayed gene expression similarities with human RLF–MYCL SCLC. Together, our studies support RLF–MYCL as the first demonstrated fusion oncogenic driver in SCLC and provide a new preclinical mouse model for the study of this subtype of SCLC.Significance:The biological and therapeutic implications of gene fusions in SCLC, an aggressive metastatic lung cancer, are unknown. Our study investigates the functional significance of the in-frame RLF–MYCL gene fusion by developing a Rlf–Mycl-driven genetically engineered mouse model and defining the impact on tumor growth and metastasis.This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2023
9. Cytokine receptor IL27RA is an NF-kB-responsive gene involved in CD38 upregulation in multiple myeloma
- Author
-
Rebecca J Brownlie, Ruth Kennedy, Erica B Wilson, Maja Milanovic, Claire F Taylor, Dapeng Wang, John Davies, Heather Elizabeth Owston, Emma J Adams, Sophie Stephenson, Rebecca Caeser, Benjamin E Gewurz, Peter V Giannoudis, Claudio Scuoppo, Dennis McGonagle, Daniel J Hodson, Reuben M Tooze, Gina M Doody, Gordon Cook, David R Westhead, and Ulf Klein
- Subjects
Hematology - Abstract
Multiple myeloma (MM) shows constitutive activation of canonical and non-canonical nuclear factor-ĸB (NF-ĸB) signaling through genetic mutations or stimuli from the tumour microenvironment (TME). A subset of MM cell lines showed dependency for cell growth and survival on the canonical NF-ĸB transcription factor RELA alone, suggesting a critical role for a RELA-mediated biological program in MM pathogenesis. Here, we determined the RELA-dependent transcriptional program in MM cell lines and found the expression of the cell surface molecules IL-27 receptor-α (IL-27Rα) and the adhesion molecule JAM2 to be responsive to RELA at the mRNA and protein levels. IL-27Rα and JAM2 were expressed on primary MM cells at higher levels than on normal long-lived plasma cells (PCs) in the bone marrow. IL-27 activated STAT1, and to a lesser extent STAT3, in MM cell lines and in PCs generated from memory B-cells in an IL-21-dependent in vitro PC-differentiation assay. Concomitant activity of IL-21 and IL-27 enhanced differentiation into PCs and increased cell-surface expression of the known STAT target gene CD38. In accordance, a subset of MM cell lines and primary MM cells cultured with IL-27 upregulated CD38 cell-surface expression, a finding with potential implications for enhancing the efficacy of CD38-directed monoclonal antibody (mAb) therapies by increasing CD38-expression on tumour cells. The elevated expression of IL-27Rα and JAM2 on MM cells compared to normal PCs may be exploited for the development of targeted therapeutic strategies that modulate the interaction of MM cells with the TME.
- Published
- 2023
10. Genetic manipulation and immortalized culture of ex vivo primary human germinal center B cells
- Author
-
Miriam Di Re, Chun Gong, Rebecca Caeser, Daniel J. Hodson, Jie Gao, Hodson, Daniel J [0000-0001-6225-2033], and Apollo - University of Cambridge Repository
- Subjects
Cell Culture Techniques ,Genomics ,Biology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,law ,medicine ,Humans ,Ex vivo expansion ,Cell Proliferation ,030304 developmental biology ,B-Lymphocytes ,0303 health sciences ,Germinal center ,Germinal Center ,medicine.disease ,In vitro ,3. Good health ,Lymphoma ,Genetic Techniques ,Cancer research ,Suppressor ,CRISPR-Cas Systems ,Gene Deletion ,030217 neurology & neurosurgery ,Ex vivo - Abstract
Next-generation sequencing has transformed our knowledge of the genetics of lymphoid malignancies. However, limited experimental systems are available to model the functional effects of these genetic changes and their implications for therapy. The majority of mature B-cell malignancies arise from the germinal center (GC) stage of B-cell differentiation. Here we describe a detailed protocol for the purification and ex vivo expansion of primary, nonmalignant human GC B cells. We present methodology for the high-efficiency transduction of these cells to enable combinatorial expression of putative oncogenes. We also describe alternative approaches for CRISPR-Cas9-mediated deletion of putative tumor suppressors. Mimicking genetic changes commonly found in lymphoid malignancies leads to immortalized growth in vitro, while engraftment into immunodeficient mice generates genetically customized, synthetic models of human lymphoma. The protocol is simple and inexpensive and can be implemented in any laboratory with access to standard cell culture and animal facilities. It can be easily scaled up to enable high-throughput screening and thus provides a versatile platform for the functional interrogation of lymphoma genomic data.
- Published
- 2021
11. Abstract 5214: Expanding the spectrum of germline-driven cancers by leveraging population-scale targeted tumor and normal sequencing
- Author
-
Miika Mehine, Rebecca Caeser, Yelena Kemel, Daniel Muldoon, Sebastià Franch-Expósito, A. Rose Brannon, Aijazuddin Syed, Ozge Ceyhan-Birsoy, Maksym Misyura, Panieh Terraf, David B. Solit, Marc Ladanyi, Kenneth Offit, Zsofia K. Stadler, Diana L. Mandelker, Yonina R. Murciano-Goroff, Charles M. Rudin, Michael F. Berger, and Chaitanya Bandlamudi
- Subjects
Cancer Research ,Oncology - Abstract
Large case-control and familial studies have established clear cancer-specific risk profiles for several key cancer predisposition genes (CPGs). For example, germline pathogenic variants (PVs) in BRCA1/2 (gBRCA) are associated with increased risk for developing breast, ovarian, pancreatic, and prostate cancers. However, the extent to which gBRCA mutations are involved in mediating the tumorigenesis of other cancer types remains challenging to characterize. We hypothesized that integrating orthogonal features such as selection for biallelic inactivation of the PVs and depletion of canonical somatic drivers among the carriers can enrich the signal for identifying novel gene and cancer type associations. We then extend this framework to identify novel CPGs as well as to understand how tumors arise in patients with PVs in oncogenes. To study this, we leveraged the prospective MSK-IMPACT matched tumor-normal sequencing cohort of 49,291 patients across 77 major cancer types. We study 90 well-known CPGs as well as >300 cancer genes not previously associated with cancer predisposition. Overall, 8% (N=3,964) of patients harbored a PV in high or moderate penetrance CPGs. We identified 90 gene and cancer type associations with enrichment for biallelic inactivation (q Among carriers of PVs in oncogenes, we observe two possible mechanisms of first somatic hit towards malignant transformation. We find enrichment for copy number gain or copy neutral loss of heterozygosity of the germline PV in thyroid cancers with a PV in RET. We also find that lung cancers with a germline PV in EGFR frequently developed additional somatic point mutations located in cis with the PV. Investigating genes with no prior association with germline predisposition to cancer, we find evidence for KEAP1 and CIC as likely novel CPGs. Lung (n=8) and thyroid (n=4) cancers with deleterious germline variants in KEAP1 were characterized by loss of the wild-type allele, co-occurring somatic STK11 mutations, and depletion of canonical drivers such as EGFR. We also found biallelic loss of CIC in two patients with Neuroblastoma, each carrying a different germline loss-of-function mutation in CIC. Both tumors were also negative for MYCN and ALK defects. Collectively, our findings expand our understanding of cancer predisposition in cancer, shed new insights into how tumors arise in germline carriers, and provide a framework for identifying new CPGs using population scale tumor-normal paired clinical sequencing data. Citation Format: Miika Mehine, Rebecca Caeser, Yelena Kemel, Daniel Muldoon, Sebastià Franch-Expósito, A. Rose Brannon, Aijazuddin Syed, Ozge Ceyhan-Birsoy, Maksym Misyura, Panieh Terraf, David B. Solit, Marc Ladanyi, Kenneth Offit, Zsofia K. Stadler, Diana L. Mandelker, Yonina R. Murciano-Goroff, Charles M. Rudin, Michael F. Berger, Chaitanya Bandlamudi. Expanding the spectrum of germline-driven cancers by leveraging population-scale targeted tumor and normal sequencing. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5214.
- Published
- 2023
12. Abstract 6127: MDM2 inhibition in combination with MEK inhibition in pre-clinical models of lung adenocarcinomas with MDM2 amplification
- Author
-
Arielle Elkrief, Vladimir Markov, Álvaro Quintanal-Villalonga, Rebecca Caeser, Pawel Sobczuk, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Charles M. Rudin, Igor Odintsov, and Romel Somwar
- Subjects
Cancer Research ,Oncology - Abstract
The eventual development of resistance to single-agent targeted therapies in lung adenocarcinomas (LUAD) is inevitable, and new strategies are needed. We hypothesize that combination therapies aimed at a known driver and a distinct targetable alteration could prolong time on oral targeted therapy. In an analysis of 7636 patients with LUAD who underwent MSK-IMPACT large panel NGS testing, 5.5% (416/7636) harbored MDM2 amplification (MDM2amp), a known mechanism of TP53 inactivation. MDM2amp was over-represented among tumors with alterations in METex14 (34.4%, p Citation Format: Arielle Elkrief, Vladimir Markov, Álvaro Quintanal-Villalonga, Rebecca Caeser, Pawel Sobczuk, Emily Cheng, Alexander Drilon, Gregory J. Riely, William W. Lockwood, Elisa de Stanchina, Charles M. Rudin, Igor Odintsov, Romel Somwar. MDM2 inhibition in combination with MEK inhibition in pre-clinical models of lung adenocarcinomas with MDM2 amplification [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6127.
- Published
- 2023
13. BCL6-Mediated Escape from Negative Selection Enables RAS-Driven B-Cell Transformation
- Author
-
Lai N. Chan, Christian Hurtz, Mark E. Robinson, Etienne Leveille, Huimin Geng, Rebecca Caeser, Liang Xu, Kohei Kume, Gang Xiao, and Markus Müschen
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
14. Abstract 90: STAT3-driven MAPK activation represents a therapeutic vulnerability in ASCL1 high SCLC
- Author
-
Rebecca Caeser, Christopher Hulton, Emily Costa, Vidushi Durani, Megan Little, Xiaoping Chen, Sam E. Tischfield, Marina Asher, Faruk Erdem Kombak, Shweta S. Chavan, Nisargbhai S. Shah, Metamia Ciampricotti, Elisa de Stanchina, John T. Poirier, Charles M. Rudin, and Triparna Sen
- Subjects
Cancer Research ,Oncology - Abstract
Background Small cell lung cancer (SCLC) is an aggressive high grade neuroendocrine tumor (Hann et al. 2019; Bernhardt et al. 2016). Whilst MAPK mutations can be found in roughly 30% of human cancers (Schubbert et al. 2007) including non-small cell lung cancer (NSCLC), genomic and proteomic analyses have indicated suppression of MAPK pathway activity in SCLC (Cerami et al. 2012; Gao et al. 2013). Previous attempts to determine whether this might be therapeutically important (Ravi et al, 1998; Cristea et al. 2016/2020) have had conflicting conclusions. SCLC has recently been defined by the relative expression of four major transcriptional regulators (ASCL1, NEUROD1, POU2F3, YAP1) (Rudin et al., 2019). In this study we aimed to elucidate the effect of MAPK activation in these different SCLC subtypes and explore its therapeutic vulnerability. Results ASCL1-, NEUROD1- and POU2F3- driven SCLC cell lines were transduced with a doxycycline-inducible vector for expression of MEKDDS217D/S221D (MEK1). Activation through MEK1 in ASCL1-driven SCLC cell lines resulted in a significant decrease in cell growth over 9 days. This was associated with G2 cell-cycle arrest and senescence. Expression of MEK1 in the cells of other SCLC subtypes and NSCLC failed to show any appreciable changes in cell growth. Remarkably, athymic mice injected with a MEK1 expressing ASCL1-driven cell line showed significantly slower tumor formation and longer survival than when MEK1 was not expressed. Notably, we observed the opposite when MEK1 expressing NEUROD1-driven cells were injected into athymic mice. We observed strong upregulation of negative feedback regulators DUSP6 and SPRY2 upon MAPK activation which has previously been described in solid tumors and pre-B ALL (Courtois-Cox et al 2006; Shojaee et al. 2015). Interestingly, phosphokinase array demonstrated that, almost exclusively, STAT3 through phosphorylation at S727 was strongly upregulated in the ASCL1-driven subtype after MEK1 expression. This prompted us to look at other regulators of the JAK-STAT pathway and found an increase in phosphorylation of the inhibitory phosphatase PTPN6 (SHP1) in the ASCL1-driven subtype and no STAT5 phosphorylation in any of the subtypes. We next examined whether these cells were sensitive to STAT3 inhibition. Upon treatment with a STAT3 inhibitor, stattic, ASCL1-driven SCLC cells reached their IC50 after 3-5 days in comparison to 9 days for other SCLC subtypes. MEK inhibition through PD0325901 rescued growth inhibition upon MAPK activation and was further associated with a decrease in DUSP6, SPRY2 and pS727 STAT3. JAK1/2 inhibition through ruxolitinib had no effect on cell viability. Summary These findings suggest that ASCL1-driven SCLC in vitro and in vivo is sensitive to activation of MAPK signaling. Our data provides additional understanding of SCLC biology and its complex signaling networks and potential subtype-specific drug susceptibilities. Citation Format: Rebecca Caeser, Christopher Hulton, Emily Costa, Vidushi Durani, Megan Little, Xiaoping Chen, Sam E. Tischfield, Marina Asher, Faruk Erdem Kombak, Shweta S. Chavan, Nisargbhai S. Shah, Metamia Ciampricotti, Elisa de Stanchina, John T. Poirier, Charles M. Rudin, Triparna Sen. STAT3-driven MAPK activation represents a therapeutic vulnerability in ASCL1 high SCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 90.
- Published
- 2022
15. Abstract 6238: Profiling of the circulating cell-free DNA methylome for detection and subtyping of small cell lung cancers
- Author
-
Dominic G. Rothwell, Francesca Chemi, Simon Pearce, Alex Clipson, Steven Hill, Alicia Marie Conway, Sophie Richardson, Katarzyna Murat, Rebecca Caeser, Jacklynn Egger, John T. Poirier, Alastair Kerr, Fiona Blackhall, Charles M. Rudin, and Caroline Dive
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Small-cell lung cancer (SCLC) is a high-grade neuroendocrine carcinoma characterised by high proliferation rate and early, rapid metastatic spread. Although SCLC is treated as a homogenous disease, recent studies revealed morphologic and transcriptomic heterogeneity with several molecular subtypes described based on predominant transcription factor expression (ASCL1, NEUROD1, ATOH1, POU2F3, YAP1) (Rudin et al., 2019; Simpson et al., 2020) which in preclinical studies exhibit differing vulnerabilities raising the potential of stratified therapy. DNA methylation is also thought be an important regulator of SCLC biology (Gazdar et al., 2017) and epigenetically distinct subtypes derived from SCLC primary tumour samples reported (Poirier et al., 2015). Here, we developed a robust workflow for genome-wide DNA methylation profiling to examine the potential use of cfDNA methylation profiling for detection and subtyping of SCLC. Results: To evaluate SCLC genome-wide DNA methylation patterns we employed a bisulfite-free enrichment-based approach (T7-MBD-seq). We tested this approach on tissue samples from preclinical models and from normal lung (n=110) and on cfDNA samples from both patients with SCLC and from non-cancer controls (n=157). Methylation profiles from preclinical models (patient-derived xenografts (PDX) and CTC derived explant (CDX) models) were comparable to previously described methylation patterns from SCLC primary tumours and were recapitulated in patients’ cfDNA samples. A tumour/normal classifier, based on 4,061 genomic regions detected as being hypermethylated in SCLC preclinical models, correctly assigned 93% and 100% cfDNA samples from patients with limited and extensive stage SCLC respectively, with a statistically significant correlation of prediction scores with disease stage (P=0.0076). Finally, to determine whether cfDNA methylation profiling could subtype SCLC patients, we built a subtype classifier, based on methylation signatures derived from 59 established SCLC cell lines. We applied the classifier to cfDNA samples from 56 patients and 10/11 with known subtypes (identified from a donor matched CDX model) were correctly classified. Overall, 73% of cfDNA samples were classified as ASCL1, 13% were classified as NEUROD1 and 14% were classified as being double negative with the distribution of the subtypes correlating closely to previously published IHC data from SCLC tissue samples (Baine et al., 2020). Conclusions: Our data reveal two potential clinical utilities of cfDNA methylation profiling; a universally applicable liquid biopsy approach for more sensitive detection and monitoring of SCLC and molecular subtyping to ease the path to future clinical trials of subtype stratified treatments for patients with SCLC. Citation Format: Dominic G. Rothwell, Francesca Chemi, Simon Pearce, Alex Clipson, Steven Hill, Alicia Marie Conway, Sophie Richardson, Katarzyna Murat, Rebecca Caeser, Jacklynn Egger, John T. Poirier, Alastair Kerr, Fiona Blackhall, Charles M. Rudin, Caroline Dive. Profiling of the circulating cell-free DNA methylome for detection and subtyping of small cell lung cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 6238.
- Published
- 2022
16. MAPK pathway activation selectively inhibits ASCL1-driven small cell lung cancer
- Author
-
John T. Poirier, Christopher H. Hulton, Shweta S. Chavan, Sam E. Tischfield, Marina Asher, Xiaoping Chen, Triparna Sen, Nisargbhai S. Shah, Metamia Ciampricotti, Emily A. Costa, Elisa de Stanchina, Rebecca Caeser, Vidushi Durani, Charles M. Rudin, Faruk Erdem Kombak, and Megan Little
- Subjects
MAPK/ERK pathway ,Senescence ,Cell biology ,Multidisciplinary ,Chemistry ,Molecular biology ,Science ,Cell ,medicine.disease ,Article ,respiratory tract diseases ,ASCL1 ,Biological sciences ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,Cancer research ,Signal transduction ,Protein kinase A ,Lung cancer ,neoplasms - Abstract
Summary Activation of mitogenic signaling pathways is a common oncogenic driver of many solid tumors including lung cancer. Although activating mutations in the mitogen-activated protein kinase (MAPK) pathway are prevalent in non-small cell lung cancers, MAPK pathway activity, counterintuitively, is relatively suppressed in the more aggressively proliferative small cell lung cancer (SCLC). Here, we elucidate the role of the MAPK pathway and how it interacts with other signaling pathways in SCLC. We find that the most common SCLC subtype, SCLC-A associated with high expression of ASCL1, is selectively sensitive to MAPK activation in vitro and in vivo through induction of cell-cycle arrest and senescence. We show strong upregulation of ERK negative feedback regulators and STAT signaling upon MAPK activation in SCLC-A lines. These findings provide insight into the complexity of signaling networks in SCLC and suggest subtype-specific mitogenic vulnerabilities., Graphical abstract, Highlights • MAPK activation causes cell-cycle arrest and senescence selectively in SCLC-A subtype • MAPK-induced growth inhibition is independent of NOTCH signaling • MAPK activation increases ERK negative feedback and activates STAT3 signaling, Biological sciences; Cell biology; Molecular biology
- Published
- 2021
17. Rlf-Mycl Gene Fusion Drives Tumorigenesis and Metastasis in a Mouse Model of Small Cell Lung Cancer
- Author
-
Triantafyllia R. Karakousi, William M. Rideout, Allison Richards, Andrea Ventura, Mark T.A. Donoghue, Janneke E. Jaspers, Triparna Sen, Elliot H. Akama-Garren, Trudy G. Oliver, Tyler Jacks, Faruk Erdem Kombak, Anastasia-Maria Zavitsanou, John T. Poirier, Metamia Ciampricotti, Álvaro Quintanal-Villalonga, Francisco J. Sánchez-Rivera, Viola Allaj, P. Manoj, Danilo Maddalo, Thales Papagiannakopoulos, Rebecca Caeser, Emily A. Costa, Angeliki Karatza, Kyle B. Spainhower, and Charles M. Rudin
- Subjects
Lung Neoplasms ,Somatic cell ,Carcinogenesis ,Telomere-Binding Proteins ,Genes, myc ,Biology ,medicine.disease_cause ,Article ,Metastasis ,Fusion gene ,Proto-Oncogene Proteins c-myc ,Mice ,Cell Line, Tumor ,Gene expression ,medicine ,CRISPR ,Animals ,neoplasms ,Cas9 ,medicine.disease ,Small Cell Lung Carcinoma ,humanities ,respiratory tract diseases ,Oncology ,Tumor progression ,Cancer research ,Gene Fusion - Abstract
Small cell lung cancer (SCLC) has limited therapeutic options and an exceptionally poor prognosis. Understanding the oncogenic drivers of SCLC may help define novel therapeutic targets. Recurrent genomic rearrangements have been identified in SCLC, most notably an in-frame gene fusion between RLF and MYCL found in up to 7% of the predominant ASCL1-expressing subtype. To explore the role of this fusion in oncogenesis and tumor progression, we used CRISPR/Cas9 somatic editing to generate a Rlf–Mycl-driven mouse model of SCLC. RLF–MYCL fusion accelerated transformation and proliferation of murine SCLC and increased metastatic dissemination and the diversity of metastatic sites. Tumors from the RLF–MYCL genetically engineered mouse model displayed gene expression similarities with human RLF–MYCL SCLC. Together, our studies support RLF–MYCL as the first demonstrated fusion oncogenic driver in SCLC and provide a new preclinical mouse model for the study of this subtype of SCLC. Significance: The biological and therapeutic implications of gene fusions in SCLC, an aggressive metastatic lung cancer, are unknown. Our study investigates the functional significance of the in-frame RLF–MYCL gene fusion by developing a Rlf–Mycl-driven genetically engineered mouse model and defining the impact on tumor growth and metastasis. This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2021
18. Should WEE(1) CHK(1) in on the FAM(122A)ily?
- Author
-
Rebecca Caeser and Triparna Sen
- Subjects
animal structures ,Lung Neoplasms ,Cell Cycle Proteins ,Biology ,environment and public health ,Article ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Humans ,CHEK1 ,Phosphorylation ,Cell Cycle Protein ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Kinase ,Cell Biology ,enzymes and coenzymes (carbohydrates) ,Cell culture ,embryonic structures ,Checkpoint Kinase 1 ,Cancer research ,Non small cell ,biological phenomena, cell phenomena, and immunity ,Protein Kinases ,030217 neurology & neurosurgery - Abstract
While effective anti-cancer drugs targeting the CHK1 kinase are advancing in the clinic, drug resistance is rapidly emerging. Here, we demonstrate that CRISPR-mediated knockout of the little-known gene FAM122A confers cellular resistance to CHK1 inhibitors and cross-resistance to ATR inhibitors. Knockout of FAM122A results in activation of PP2A-B55α, a phosphatase which dephosphorylates the WEE1 protein and rescues WEE1 from Ubiquitin-mediated degradation. The resulting increase in WEE1 protein expression reduces replication stress, activates the G2/M checkpoint, and confers cellular resistance to CHK1 inhibitors. Interestingly, in tumor cells with oncogene-driven replication stress, CHK1 can directly phosphorylate FAM122A, leading to activation of the PP2A-B55α phosphatase and increased WEE1 expression. A combination of a CHK1 inhibitor plus a WEE1 inhibitor can overcome CHK1 inhibitor resistance of these tumor cells, thereby enhancing anti-cancer activity. The FAM122A expression level in a tumor cell can serve as a useful biomarker for predicting CHK1 inhibitor sensitivity or resistance.
- Published
- 2020
19. Functional interplay of Epstein-Barr virus oncoproteins in a mouse model of B cell lymphomagenesis
- Author
-
Timm Weber, Anna Jauch, Rebecca Caeser, Jonathan Ronen, Tomoharu Yasuda, Daniel J. Hodson, Kristian Unger, Ulrike Sack, Altuna Akalin, Klaus Rajewsky, Van Trung Chu, Thomas Sommermann, Xun Li, Jingwei Zhang, and Tristan Wirtz
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Lymphoma, B-Cell ,Transforming virus ,Plasma Cells ,Primary Cell Culture ,Biology ,medicine.disease_cause ,Virus ,Viral Matrix Proteins ,03 medical and health sciences ,Mice ,Viral Proteins ,0302 clinical medicine ,Immune system ,Antigen ,hemic and lymphatic diseases ,Cell Line, Tumor ,Plasma cell differentiation ,medicine ,otorhinolaryngologic diseases ,Animals ,Humans ,B cell ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Cell Differentiation ,Fibroblasts ,Biological Sciences ,Cell Transformation, Viral ,Epstein–Barr virus ,3. Good health ,DNA-Binding Proteins ,Transformation (genetics) ,stomatognathic diseases ,Disease Models, Animal ,medicine.anatomical_structure ,Epstein-Barr Virus Nuclear Antigens ,030220 oncology & carcinogenesis ,Cancer research ,Trans-Activators - Abstract
Epstein-Barr virus (EBV) is a B cell transforming virus that causes B cell malignancies under conditions of immune suppression. EBV orchestrates B cell transformation through its latent membrane proteins (LMPs) and Epstein-Barr nuclear antigens (EBNAs). We here identify secondary mutations in mouse B cell lymphomas induced by LMP1, to predict and identify key functions of other EBV genes during transformation. We find aberrant activation of early B cell factor 1 (EBF1) to promote transformation of LMP1-expressing B cells by inhibiting their differentiation to plasma cells. EBV EBNA3A phenocopies EBF1 activities in LMP1-expressing B cells, promoting transformation while inhibiting differentiation. In cells expressing LMP1 together with LMP2A, EBNA3A only promotes lymphomagenesis when the EBNA2 target Myc is also overexpressed. Collectively, our data support a model where proproliferative activities of LMP1, LMP2A, and EBNA2 in combination with EBNA3A-mediated inhibition of terminal plasma cell differentiation critically control EBV-mediated B cell lymphomagenesis.
- Published
- 2020
20. Genetic modification of primary human B cells to model high-grade lymphoma
- Author
-
Zelvera Usheva, George S. Vassiliou, Joanna Alicja Krupka, Maribel Lara-Chica, Chan-Sik Park, Hendrik F. P. Runge, Brian J. P. Huntly, Hyo-Kyung Pak, Philip A. Beer, Susanna L. Cooke, Rachael Bashford-Rogers, João M. L. Dias, Hesham Eldaly, Rachel Fenner, Daniel J. Hodson, Jie Gao, Annalisa Mupo, Rebecca Caeser, Miriam Di Re, Krupka, Joanna A. [0000-0003-0369-0329], Lara-Chica, Maribel [0000-0003-2908-9929], Dias, João M. L. [0000-0002-8451-3537], Runge, Hendrik F. P. [0000-0002-4282-1515], Vassiliou, George S. [0000-0003-4337-8022], Huntly, Brian J. P. [0000-0003-0312-161X], Bashford-Rogers, Rachael J. M. [0000-0002-6838-0711], Hodson, Daniel J. [0000-0001-6225-2033], Apollo - University of Cambridge Repository, Krupka, Joanna A [0000-0003-0369-0329], Dias, João ML [0000-0002-8451-3537], Runge, Hendrik FP [0000-0002-4282-1515], Vassiliou, George S [0000-0003-4337-8022], Huntly, Brian JP [0000-0003-0312-161X], Bashford-Rogers, Rachael JM [0000-0002-6838-0711], and Hodson, Daniel J [0000-0001-6225-2033]
- Subjects
0301 basic medicine ,96 ,General Physics and Astronomy ,14 ,Transduction (genetics) ,Mice ,0302 clinical medicine ,Transduction, Genetic ,immune system diseases ,hemic and lymphatic diseases ,38/23 ,CRISPR ,lcsh:Science ,B-Lymphocytes ,Multidisciplinary ,article ,BCL6 ,3. Good health ,13/31 ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Proto-Oncogene Proteins c-bcl-6 ,38/77 ,Lymphoma, Large B-Cell, Diffuse ,631/67/70 ,Science ,Tumour heterogeneity ,Genetic Vectors ,Primary Cell Culture ,13/107 ,13/106 ,13/109 ,Biology ,631/67/2329 ,General Biochemistry, Genetics and Molecular Biology ,38 ,38/91 ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cancer models ,Cell Proliferation ,Cas9 ,Germinal center ,General Chemistry ,medicine.disease ,Germinal Center ,Xenograft Model Antitumor Assays ,Coculture Techniques ,Lymphoma ,High-Throughput Screening Assays ,030104 developmental biology ,Retroviridae ,Cell culture ,13/51 ,Cancer research ,lcsh:Q ,CRISPR-Cas Systems ,Neoplasm Grading ,Diffuse large B-cell lymphoma - Abstract
Sequencing studies of diffuse large B cell lymphoma (DLBCL) have identified hundreds of recurrently altered genes. However, it remains largely unknown whether and how these mutations may contribute to lymphomagenesis, either individually or in combination. Existing strategies to address this problem predominantly utilize cell lines, which are limited by their initial characteristics and subsequent adaptions to prolonged in vitro culture. Here, we describe a co-culture system that enables the ex vivo expansion and viral transduction of primary human germinal center B cells. Incorporation of CRISPR/Cas9 technology enables high-throughput functional interrogation of genes recurrently mutated in DLBCL. Using a backbone of BCL2 with either BCL6 or MYC, we identify co-operating genetic alterations that promote growth or even full transformation into synthetically engineered DLBCL models. The resulting tumors can be expanded and sequentially transplanted in vivo, providing a scalable platform to test putative cancer genes and to create mutation-directed, bespoke lymphoma models., A dearth of adequate preclinical models to faithfully mimic diffuse large B-cell lymphoma has hampered the identification of driver genes. Here, the authors present a co-culture system that enables ex vivo expansion, viral transduction and transformation of primary human germinal center B cells.
- Published
- 2020
21. Signaling input from divergent pathways subverts malignant B-cell transformation
- Author
-
Teresa Sadras, Andreas Vogt, Seyedmehdi Shojaee, Eamon Aghania, Kadriye Nehir Cosgun, Olli Lohi, Jae-Woong Lee, David M. Weinstock, Markus Müschen, Vishal Khairnar, Gang Xiao, Rebecca Caeser, Christian Hurtz, Chun-Wei Chen, Mark E. Robinson, Matthew A. Nix, Merja Heinäniemi, Mohamed R. Ahmed, Mark A. Murakami, Arun P. Wiita, Huimin Geng, Jianjun Chen, Lai N. Chan, Chao Hong, Kohei Kume, Shai Izraeli, Gauri Deb, Petri Pölönen, and Zhengshan Chen
- Subjects
MAPK/ERK pathway ,0303 health sciences ,Mutation ,biology ,medicine.disease_cause ,BCL6 ,Malignant transformation ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Signal transduction ,Receptor ,B cell ,STAT5 ,030304 developmental biology - Abstract
Malignant transformation typically involves multiple genetic lesions whose combined activity gives rise to cancer1. Our analysis of 1,148 patient-derived B-cell leukemia (B-ALL) samples revealed that individual mutations did not promote leukemogenesis unless they converged on one single oncogenic pathway characteristic for the differentiation status of these transformed B cells. Specifically, we show here the JAK/STAT5 signaling pathway supports the developmental stage-specific expansion of pro-B ALL whereas the ERK-pathway that of pre-B ALL. Mutations that were not aligned with the central oncogenic driver would activate divergent pathways and subvert malignant transformation. Oncogenic lesions in B-ALL frequently mimic survival and proliferation signals downstream of cytokine receptors (through activation of STAT5)2-7 or the pre-B cell receptor (through activation of ERK)8-13. STAT5- (372 cases) and ERK- (386 cases) activating lesions were frequently found but only co-occurred in ∼3% (37) of cases (P=2.2E-16). Single-cell mutation and phosphoprotein analyses revealed that even in these rare cases, oncogenic STAT5- or ERK-activation were mutually exclusive and segregated to competing clones. STAT5 and ERK engaged opposing biochemical and transcriptional programs orchestrated by MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway came at the expense of the principal oncogenic driver and reversed malignant transformation. Conversely, Cre-mediated deletion of divergent pathway components triggered leukemia-initiation and accelerated development of fatal disease. Thus, persistence of divergent signaling pathways represents a powerful barrier to malignant transformation while convergence on one principal driver defines a key event during leukemia-initiation. Proof-of-concept studies in patient-derived B-ALL cells revealed that pharmacological reactivation of suppressed divergent circuits strongly synergized with direct inhibition of the principal oncogenic driver. Hence, pharmacological reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to deepen treatment responses and to overcome drug-resistance. Current treatment approaches for drug-resistant cancer are focused on drug-combinations to suppress the central oncogenic driver and multiple alternative pathways14-17. Here, we introduce a concept based on inhibition of the principal driver combined with pharmacological reactivation of divergent pathways.
- Published
- 2020
- Full Text
- View/download PDF
22. Abstract LB186: MAPK pathway activation represents a therapeutic vulnerability in ASCL1-driven SCLC
- Author
-
Nisargbhai S. Shah, Megan Little, John T. Poirier, Emily Costa, Triparna Sen, Charles M. Rudin, Vidushi Durani, Rebecca Caeser, Christopher H. Hulton, and Elisa de Stanchina
- Subjects
MAPK/ERK pathway ,YAP1 ,Cancer Research ,Cancer ,Biology ,medicine.disease ,Metastasis ,Oncology ,Downregulation and upregulation ,Apoptosis ,SPRY2 ,medicine ,Cancer research ,biology.protein ,STAT3 - Abstract
Background Lung cancer is the leading cause of cancer death, killing more people than colon, breast, and prostate cancers combined (Siegel et al. 2013). Small cell lung cancer (SCLC) is a high grade neuroendocrine tumor accounting for ~15% of all lung cancers (Hann et al. 2019). Metastasis is often found at first diagnosis, making SCLC exceptionally lethal (2-year survival <5%) (Byers et al. 2015). Whilst MAPK mutations can be found in roughly 30% of human cancers (Schubbert et al. 2007) including non-small cell lung cancer (NSCLC), genomic and proteomic analyses have indicated suppression of MAPK pathway activity in SCLC (Cerami et al. 2012; Gao et al. 2013; Wagle et al. 2018). This striking difference is not well understood and previous attempts to determine whether this might be therapeutically important (Ravi et al, 1998; Cristea et al. 2020) have had conflicting conclusions. SCLC has recently been defined by the relative expression of four major transcriptional regulators (ASCL1, NeuroD1, POU2F3, YAP1) (Rudin et al., 2019). In this study we aimed to elucidate the effect of MAPK activation in these different SCLC subtypes and explore its therapeutic vulnerability. Results We used a doxycycline-inducible vector for expression of MEKDDS217D/S221D (MEK1) in a cohort of ASCL1-, NEUROD1, and POU2F3- driven cell lines. Activation through MEK1 in ASCL1-driven SCLC cell lines resulted in a significant decrease in cell growth over 9 days. This was associated with a decrease in neuroendocrine markers ASCL1 and INSM1, a G2 cell cycle arrest and no significant increase in apoptotic cells. Expression of MEK1 in other SCLC subtypes and NSCLC failed to show any appreciable changes in cell growth. Remarkably, athymic mice injected with a MEK1 expressing ASCL1-driven cell line showed significantly slower tumor formation and longer survival than the ASCL1-driven cell line not expressing MEK1. Previous work established that hyperactivation of BRAFV600E, RAS and MYC can result in oncogene-induced senescence (Serrano et al. 1997) which is caused by upregulation of negative feedback such as SPRY2, DUSP6, ETV5 rather than MAPK pathway activation in some solid tumors and pre-B ALL (Courtois-Cox et al 2006; Shojaee et al. 2015). Similarly, we also observed strong upregulation of DUSP6, SPRY2, but not ETV5 upon MAPK activation. This was especially prominent in ASCL1-driven cell lines that changed from the normal phenotype of being in suspension to a more adherent morphology as a result of MAPK activation. Interestingly, phosphokinase array in the major subtype cell lines after MEK1 activation, demonstrated that, almost exclusively, the STAT pathways, in particular STAT3 through phosphorylation at S727 was strongly upregulated in the ASCL1-driven subtype. This prompted us to examine whether these cells were sensitive to STAT3 inhibition. Upon treatment with a STAT3 inhibitor, Stattic (1μM), ASCL1-driven SCLC cells reached their IC50 after 3-5 days in comparison to 9 days for other SCLC subtypes. NSCLC cells were resistant to STAT3 inhibition. Summary These findings suggest that ASCL1-driven SCLC in vitro and in vivo is sensitive to activation of MAPK signaling in comparison to other SCLC subtypes. Whilst activation of the MAPK pathway might seem counterintuitive to current treatment strategies that aim to inhibit oncogenic signaling, we propose the use of a STAT3 inhibitor that has shown to be effective in vitro. Citation Format: Rebecca Caeser, Christopher Hulton, Vidushi Durani, Emily Costa, Megan Little, Nisargbhai S. Shah, Elisa de Stanchina, John T. Poirier, Charles M. Rudin, Triparna Sen. MAPK pathway activation represents a therapeutic vulnerability in ASCL1-driven SCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr LB186.
- Published
- 2021
23. Signalling input from divergent pathways subverts B cell transformation
- Author
-
Zhengshan Chen, Arun P. Wiita, Seyedmehdi Shojaee, Chao Hong, Kadriye Nehir Cosgun, Gauri Deb, Mohamed A. Ahmed, Kohei Kume, Gang Xiao, Olli Lohi, Mark A. Murakami, Jae-Woong Lee, David M. Weinstock, Jianjun Chen, Mark E. Robinson, Shai Izraeli, Rebecca Caeser, Teresa Sadras, Christian Hurtz, Huimin Geng, Matthew A. Nix, Petri Pölönen, Chun-Wei Chen, Merja Heinäniemi, Andreas Vogt, Eamon Aghania, Markus Müschen, Lai N. Chan, and Vishal Khairnar
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Protein tyrosine phosphatase ,medicine.disease_cause ,Article ,Malignant transformation ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Leukemia, B-Cell ,STAT5 Transcription Factor ,Animals ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Transcription factor ,STAT5 ,Mutation ,B-Lymphocytes ,Multidisciplinary ,biology ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,Hedgehog signaling pathway ,Cell biology ,Enzyme Activation ,030104 developmental biology ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,biology.protein ,Proto-Oncogene Proteins c-bcl-6 ,Female ,Signal transduction ,Signal Transduction - Abstract
Malignant transformation of cells typically involves several genetic lesions, whose combined activity gives rise to cancer1. Here we analyse 1,148 patient-derived B-cell leukaemia (B-ALL) samples, and find that individual mutations do not promote leukaemogenesis unless they converge on one single oncogenic pathway that is characteristic of the differentiation stage of transformed B cells. Mutations that are not aligned with this central oncogenic driver activate divergent pathways and subvert transformation. Oncogenic lesions in B-ALL frequently mimic signalling through cytokine receptors at the pro-B-cell stage (via activation of the signal-transduction protein STAT5)2–4 or pre-B-cell receptors in more mature cells (via activation of the protein kinase ERK)5–8. STAT5- and ERK-activating lesions are found frequently, but occur together in only around 3% of cases (P = 2.2 × 10−16). Single-cell mutation and phospho-protein analyses reveal the segregation of oncogenic STAT5 and ERK activation to competing clones. STAT5 and ERK engage opposing biochemical and transcriptional programs that are orchestrated by the transcription factors MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway comes at the expense of the principal oncogenic driver and reverses transformation. Conversely, deletion of divergent pathway components accelerates leukaemogenesis. Thus, persistence of divergent signalling pathways represents a powerful barrier to transformation, while convergence on one principal driver defines a central event in leukaemia initiation. Pharmacological reactivation of suppressed divergent circuits synergizes strongly with inhibition of the principal oncogenic driver. Hence, reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to enhance treatment responses. Analysis of B-cell leukaemia samples reveals that oncogenic mutations do not cause malignant transformation unless they converge on the same signalling pathway, and that it may be possible clinically to combine inhibition of the principal oncogenic driver with reactivation of divergent pathways.
- Published
- 2019
24. Genetic modification of primary human B cells generates translationally-relevant models of high-grade lymphoma
- Author
-
Susanna L. Cooke, M. Lara-Chica, Joanna Alicja Krupka, Hesham Eldaly, George S. Vassiliou, Brian J. P. Huntly, Jie Gao, Zelvera Usheva, Chan-Sik Park, Annalisa Mupo, Rachael Bashford-Rogers, Di Re M, Daniel J. Hodson, João M. L. Dias, Hendrik F. P. Runge, Rebecca Caeser, Hyo-Kyung Pak, Rachel Fenner, and Phillip Beer
- Subjects
0303 health sciences ,Cas9 ,Germinal center ,Computational biology ,Biology ,medicine.disease ,BCL6 ,Lymphoma ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Cell culture ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,CRISPR ,Diffuse large B-cell lymphoma ,Gene ,030304 developmental biology - Abstract
Sequencing studies of Diffuse Large B Cell Lymphoma (DLBCL) have identified hundreds of recurrently altered genes. However, it remains largely unknown whether and how these mutations may contribute to lymphomagenesis, either individually or in combination. Existing strategies to address this problem predominantly utilize cell lines, which are limited by their initial characteristics and subsequent adaptions to prolongedin vitroculture. Here, we describe a novel co-culture system that enables theex vivoexpansion and viral transduction of primary human germinal center B cells. The incorporation of CRISPR/Cas9 technology enables high-throughput functional interrogation of genes recurrently mutated in DLBCL. Using a backbone ofBCL2with eitherBCL6orMYCwe have identified co-operating oncogenes that promote growth and survival, or even full transformation into synthetically engineered models of DLBCL. The resulting tumors can be expanded and sequentially transplantedin vivo, providing a scalable platform to test putative cancer genes and for the creation of mutation-directed, bespoke lymphoma models.
- Published
- 2019
25. Abstract 5469: RAS and STAT5 pathway lesions are mutually exclusive in B-cell malignancies through mechanisms of biochemical cross-inhibition
- Author
-
Christian Hurtz, Steven M. Kornblau, Lai N. Chan, Huimin Geng, Gang Xiao, Markus Müschen, Seyedmehdi Shojaee, and Rebecca Caeser
- Subjects
Cancer Research ,medicine.anatomical_structure ,Oncology ,biology ,Chemistry ,Cancer research ,biology.protein ,medicine ,Mutually exclusive events ,STAT5 ,B cell ,Cross inhibition - Abstract
Activation of STAT5- and RAS-signaling are segregated to early and later stages of normal B cell development, respectively. Studying B-lineage acute lymphoblastic leukemia (ALL; n=578), we found that STAT5 (e.g. BCR-ABL1, JAK2, cytokine receptors) and RAS (NRAS, KRAS, PTPN11, NF1) lesions were mutually exclusive, with only 9 cases (1.6%) carrying lesions in both pathways. Reverse phase protein array measurements revealed that phosphorylation of MEK and ERK1/2 were inversely correlated with STAT5-phosphorylation (MDACC, 1983-2007; P Citation Format: Lai N. Chan, Seyedmehdi Shojaee, Christian Hurtz, Rebecca Caeser, Gang Xiao, Huimin Geng, Steven Kornblau, Markus Muschen. RAS and STAT5 pathway lesions are mutually exclusive in B-cell malignancies through mechanisms of biochemical cross-inhibition [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5469.
- Published
- 2018
26. BCL6 Is Critical to Overcome Oncogene-Induced Senescence in RAS-Mediated B Cell Transformation
- Author
-
Markus Müschen, Gang Xiao, Seyedmehdi Shojaee, Ari Melnick, Lai N. Chan, Huimin Geng, Christian Hurtz, and Rebecca Caeser
- Subjects
MAPK/ERK pathway ,biology ,MEK inhibitor ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Leukemia ,medicine.anatomical_structure ,Ras Signaling Pathway ,immune system diseases ,Cell culture ,hemic and lymphatic diseases ,medicine ,biology.protein ,Cancer research ,Signal transduction ,B cell ,STAT5 - Abstract
Background and Hypothesis: The transcriptional repressor and proto-oncogene BCL6 is a therapeutic target in subtypes of diffuse large B cell lymphoma (DLBCL) and modulates drug-resistance in Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL; Duy et al., Nature 2011). BCL6 was shown to be a critical factor that bypasses p53-dependent senescence and thereby enables RAS-driven transformation of mouse embryonic fibroblasts (Shvarts et al., Genes Dev. 2002). Given that ~50% of pediatric ALL cases carry genetic lesions that lead to hyperactivated RAS-ERK signaling (Zhang et la., Blood 2012), we examined the role of BCL6 in RAS-driven pre-B ALL and identified a novel mechanism by which RAS-ERK signaling can mediate BCL6 expression. Results: Using a doxycycline-inducible TetOn- NRASG12D vector system, we found that inducible activation of RAS-ERK signaling strongly upregulated BCL6 expression at both the mRNA (~350-fold) and protein (~50-fold) levels in murine pre-B cells. Increases in BCL6 expression were abrogated upon treatment with a MEK inhibitor (PD325901). In addition, Cre-mediated deletion of Mapk1 suppressed upregulation of BCL6 expression upon imatinib treatment in BCR-ABL1-driven pre-B ALL cells. These findings suggested that elevated expression of BCL6 is a consequence of ERK activation. Previously, we demonstrated that BCL6 expression is negatively regulated by STAT5 in BCR-ABL1 pre-B ALL (Duy et al., Nature 2011). Interestingly, oncogenic NRASG12D inhibited phosphorylation of STAT5-Y694 by activating the inhibitory protein tyrosine phosphatase Ptpn6. Cre-mediated deletion of Ptpn6 induced STAT5 activity. Furthermore, loss of Ptpn6 function abrogated upregulation of BCL6 expression induced by imatinib in BCR-ABL1 pre-B ALL. Taken together, RAS-ERK signaling induces BCL6 expression by suppressing STAT5 activity. To directly test the role of BCL6 in RAS-transformed pre-B ALL, we generated a novel mouse model for inducible Cre-mediated deletion of Bcl6 exons 5-10, flanked by loxP sites. Inducible deletion of Bcl6 in NRASG12D-transformed pre-B ALL cells led to rapid depletion from the cell culture and reduced colony forming ability in vitro. These findings suggested that BCL6 is required for maintenance of fully established RAS-transformed ALL. Notably, we found that initiation of NRASG12D-driven leukemia in vivo depends on BCL6 as NRASG12D ALL failed to give rise to leukemia in the absence of Bcl6 in transplant recipient mice. Studying a diagnostic (KRAS wild-type) and a relapsed (KRASG12V) sample from one pre-B ALL patient revealed increased BCL6 expression in KRASG12V relapsed ALL cells. In addition, selective sensitivity to PD325901 and a retro inverso BCL6 peptide inhibitor (RI-BPI) was observed in KRASG12V relapsed ALL cells. Finally, RI-BPI prolonged overall survival of recipient mice transplanted with KRASG12V relapsed ALL cells in vivo. Conclusions: In summary, we demonstrated a novel mechanism by which oncogenic RAS signaling induces expression of BCL6, and showed that BCL6 is critical for RAS-driven transformation in pre-B ALL. Importantly, ALL clones often acquire drug resistance and activating mutations in the RAS pathway (Bhojwani and Pui, Lancet Oncol. 2013). Our findings suggest that pharmacological inhibition of BCL6 may provide a novel therapeutic avenue to overcome drug-resistance and prevent leukemia relapse after initial remission in RAS-driven ALL. Disclosures Melnick: Janssen: Research Funding.
- Published
- 2016
27. ATR inhibition activates cancer cell cGAS/STING-interferon signaling and promotes antitumor immunity in small-cell lung cancer.
- Author
-
Hirokazu Taniguchi, Chakraborty, Subhamoy, Nobuyuki Takahashi, Banerjee, Avisek, Rebecca Caeser, Zhan, Yingqian A., Tischfield, Sam E., Chow, Andrew, Nguyen, Evelyn M., Villalonga, Álvaro Quintanal, Manoj, Parvathy, Shah, Nisargbhai S., Rosario, Samantha, Hayatt, Omar, Rui Qu, de Stanchina, Elisa, Chan, Joseph, Hiroshi Mukae, Thomas, Anish, and Rudin, Charles M.
- Subjects
- *
PROGRAMMED death-ligand 1 , *IMMUNE checkpoint proteins , *ATAXIA telangiectasia , *T cells , *ANIMAL models in research - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Erk Negative Feedback Control Enables Pre-B Cell Transformation and Represents a Therapeutic Target in Acute Lymphoblastic Leukemia
- Author
-
Maike Buchner, Wolf-Karsten Hofmann, Stephen P. Hunger, Seyedmehdi Shojaee, Lars Klemm, Jeffery D. Molkentin, Yihua Qiu, Steven M. Kornblau, Lai N. Chan, Rebecca Caeser, Nianxiang Zhang, Markus Müschen, Ari Melnick, H. Phillip Koeffler, Srividya Swaminathan, Huimin Geng, Elisabeth Paietta, Cheryl L. Willman, Kevin R. Coombes, Christian Hurtz, and Eugene Park
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cell ,Cell Transformation ,Transgenic ,Malignant transformation ,Mice ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Host cell factor C1 ,Pediatric ,0303 health sciences ,Intracellular Signaling Peptides and Proteins ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Cell biology ,DNA-Binding Proteins ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Development of treatments and therapeutic interventions ,Programmed cell death ,MAP Kinase Signaling System ,Childhood Leukemia ,Pediatric Cancer ,Molecular Sequence Data ,Oncology and Carcinogenesis ,Mice, Transgenic ,Antineoplastic Agents ,Biology ,Protein Serine-Threonine Kinases ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Rare Diseases ,Dual Specificity Phosphatase 6 ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Transcription factor ,B cell ,030304 developmental biology ,Neoplastic ,Neurosciences ,Membrane Proteins ,Cell Biology ,SPRY2 ,Host Cell Factor C1 ,Transcription Factors - Abstract
© 2015 Elsevier Inc.. Studying mechanisms of malignant transformation of human pre-B cells, we found that acute activation ofoncogenes induced immediate cell death in the vast majority of cells. Few surviving pre-B cell clones had acquired permissiveness to oncogenic signaling by strong activation of negative feedback regulation ofErk signaling. Studying negative feedback regulation of Erk in genetic experiments at three different levels,we found that Spry2, Dusp6, and Etv5 were essential for oncogenic transformation in mouse modelsfor pre-B acute lymphoblastic leukemia (ALL). Interestingly, a small molecule inhibitor of DUSP6 selectively induced cell death in patient-derived pre-B ALL cells and overcame conventional mechanisms of drug-resistance. Shojaee etal. show that successful transformation of pre-B cells to pre-B acute lymphoblastic leukemia (ALL) requires negative feedback regulation of Erk signaling and inhibiting this feedback selectively kills pre-B ALL cells, suggesting negative feedback regulation of oncogenes as a vulnerability in human ALL.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.