20 results on '"Ram Nambiar"'
Search Results
2. Supplementary Figure 4 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Multispectral immunofluorescence. Number of cells positive for exhaustion (PD-L1, TIM3, LAG3, CTLA4) and activation markers (OX40) in baseline tumor specimens (A); Quantitation of signals per specimen (B). There are more PDL1 positive tumor cell in the pre-treatment metastatic site biopsy. While there are more activated, partially activated T cell-1 and exhausted T cell-1 in the primary tumor specimens (*p
- Published
- 2023
3. Data from Functional Determinants of Cell Cycle Plasticity and Sensitivity to CDK4/6 Inhibition
- Author
-
Erik S. Knudsen, Agnieszka K. Witkiewicz, Ram Nambiar, Paris Vail, and Vishnu Kumarasamy
- Abstract
Intrinsic or acquired resistance to clinically approved CDK4/6 inhibitors has emerged as a major obstacle that hinders their utility beyond ER+ breast cancer. In this study, CDK4/6-dependent and -resistant models were employed to identify functional determinants of response to pharmacologic CDK4/6 inhibitors. In all models tested, the activation of RB and inhibition of CDK2 activity emerged as determinants of sensitivity. While depleting CDK4 and 6 was sufficient to limit proliferation in specific resistance settings, RB loss rendered cells completely independent of these kinases. The main downstream target in this context was the activation status of CDK2, which was suppressed with CDK4/6 inhibition in an RB-dependent fashion. Protein levels of p27 were associated with plasticity/rigidity of the cell cycle and correlated with sensitivity to CDK4/6 inhibition. Exogenous overexpression and pharmacologic induction of p27 via inhibition of SKP2 and targeting the MEK/ERK pathway enhanced the cytostatic effect of CDK4/6 inhibitors. Mice bearing ER+ xenografts displayed a durable antitumor response to palbociclib; however, over the course of treatment, few cells retained RB phosphorylation, which was associated with limited p27 protein levels as determined by multispectral imaging. Similarly, combination treatment of palbociclib with a MEK inhibitor in pancreatic cancer PDX models upregulated p27 and further enhanced the in vivo tumor response to palbociclib. Collectively, these results suggest that the cell cycle plasticity, which enables tumor models to evade palbociclib-mediated activation of RB, could be targeted using a clinically applicable CDK2 inhibitor.Significance:This work provides a mechanistic insight toward understanding the functional roles of multiple cell cycle regulators that drive plasticity and sensitivity to CDK4/6 inhibition.
- Published
- 2023
4. Supplementary Data from Functional Determinants of Cell Cycle Plasticity and Sensitivity to CDK4/6 Inhibition
- Author
-
Erik S. Knudsen, Agnieszka K. Witkiewicz, Ram Nambiar, Paris Vail, and Vishnu Kumarasamy
- Abstract
Supplementary information
- Published
- 2023
5. Supplementary Figure 1 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Consort Diagram.
- Published
- 2023
6. Supplementary Figure 3 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Intratumoral NK cells (baseline) by change in tumor size (A) and change in CEA (B).
- Published
- 2023
7. Supplementary Table 2 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Correlate of baseline and on-treatment changes in intratumoral immune cells (flow cytometry) with clinical endpoints (Supplementary Table 2A), immune checkpoint expression by multispectral immunofluorescence in tumor microenvironment (Supplementary Table 2B) and correlation with clinical endpoints (Supplementary Table 2C), immune cells (flow cytometry) in peripheral blood samples at baseline and on treatment (Supplementary Table 2D) and correlation with clinical outcomes (Supplementary Table 2E).
- Published
- 2023
8. Supplementary Figure 5 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Changes in peripheral blood immune cell populations on treatment.
- Published
- 2023
9. Data from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Purpose:We evaluated the antitumor efficacy of cetuximab in combination with pembrolizumab in patients with RAS wild-type (RASwt), metastatic colorectal adenocarcinoma (mCRC).Patients and Methods:In this phase Ib/II study, cetuximab was combined with pembrolizumab in patients with RASwt mCRC with ≥ one prior line of therapy for advanced disease. We analyzed baseline on-treatment tumor tissues for changes in the tumor microenvironment (TME), using flow cytometry and multispectral immunofluorescence.Results:Forty-four patients were evaluable for efficacy. The study was negative for the primary efficacy endpoint [overall response rate: 2.6%, 6-month progression-free survival (PFS): 31%; P = 0.52]. Median PFS was 4.1 months [95% confidence interval (CI): 3.9–5.5 months]. No increase in adverse effects was identified. We observed favorable immunomodulation with 47% increase in the number of intratumoral CTLs posttreatment (P = 0.035). These changes were more pronounced in patients with tumor shrinkage (P = 0.05). The TME was characterized by high numbers of TIM3+ and CTLA4+ cells; there were few activated OX40+ cells. PD-L1 expression was higher in pretreatment tumor cells from metastatic sites versus primary tumor samples (P < 0.05). Higher numbers of PD-L1+ tumor cells at baseline were associated with tumor shrinkage (P = 0.04). Analysis of immune populations in the blood demonstrated decreases in PD-1+ memory effector cells (P = 0.04) and granulocytic myeloid-derived suppressor cells (P = 0.03), with simultaneous increases in CD4+/CTLA4+ cells (P = 0.01).Conclusions:The combination of cetuximab and pembrolizumab is inactive in patients with RASwt mCRC, despite its partial local immunologic efficacy. Further development of immuno-oncology combinations with enhanced efficacy and/or targeting additional or alternative immune checkpoints merits investigation.
- Published
- 2023
10. Supplementary Table 1 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Adverse events with maximum grade seen
- Published
- 2023
11. Supplementary Figure 2 from Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Patrick M. Boland, Renuka Iyer, Pawel Kalinski, Jennifer A. Jurcevic, Karen A. Hicks, Kristopher Attwood, Alan D. Hutson, Chong Wang, Scott I. Abrams, Jason B. Muhitch, Erik S. Knudsen, Hanna R. Rosenheck, Ram Nambiar, Hans Minderman, Orla Maguire, Agnieszka K. Witkiewicz, Joel Saltzman, Sarbajit Mukherjee, David L. Bajor, and Christos Fountzilas
- Abstract
Change in CEA over time.
- Published
- 2023
12. Pan-cancer molecular analysis of the RB tumor suppressor pathway
- Author
-
Dominic J. Smiraglia, David W. Goodrich, Spencer Rosario, Erik S. Knudsen, Ram Nambiar, and Agnieszka K. Witkiewicz
- Subjects
0301 basic medicine ,Tumor suppressor gene ,Retinal Neoplasms ,Ubiquitin-Protein Ligases ,Medicine (miscellaneous) ,Locus (genetics) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,Gene expression ,Databases, Genetic ,medicine ,Biomarkers, Tumor ,Cancer genomics ,Humans ,Tumour-suppressor proteins ,Gene ,Cancer genetics ,lcsh:QH301-705.5 ,Cell Proliferation ,Pan cancer ,Chromosomes, Human, Pair 13 ,Retinoblastoma ,Gene Expression Profiling ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Prognosis ,Molecular analysis ,Gene Expression Regulation, Neoplastic ,Retinoblastoma Binding Proteins ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Cancer research ,Disease Progression ,Suppressor ,General Agricultural and Biological Sciences ,Transcriptome ,Signal Transduction - Abstract
The retinoblastoma tumor suppressor gene (RB1) plays a critical role in coordinating multiple pathways that impact cancer initiation, disease progression, and therapeutic responses. Here we probed molecular features associated with the RB-pathway across 31 tumor-types. While the RB-pathway has been purported to exhibit multiple mutually exclusive genetic events, only RB1 alteration is mutually exclusive with deregulation of CDK4/6 activity. An ER+ breast cancer model with targeted RB1 deletion was used to identify signatures of CDK4/6 activity and RB-dependency (CDK4/6-RB integrated signature). This signature was prognostic in tumor-types with gene expression features indicative of slower growth. Single copy loss on chromosome 13q encompassing the RB1 locus is prevalent in many cancers, yielding reduced expression of multiple genes in cis, and is inversely related to the CDK4/6-RB integrated signature supporting a cause-effect relationship. Genes that are positively and inversely correlated with the CDK4/6-RB integrated signature define new tumor-specific pathways associated with RB-pathway activity., Erik Knudsen et al. present a pan-cancer analysis of the RB tumor suppressor protein pathway in 31 tumor types. They find that pathway deregulation is multi-faceted with context dependent association with survival. However, gene expression features are surprisingly invariant and support new therapeutic targets.
- Published
- 2020
13. Phase Ib/II Study of Cetuximab plus Pembrolizumab in Patients with Advanced RAS Wild-Type Colorectal Cancer
- Author
-
Joel N. Saltzman, Karen A. Hicks, Erik S. Knudsen, Hans Minderman, Scott I. Abrams, Patrick M Boland, Chong Wang, David L. Bajor, Orla Maguire, Alan D. Hutson, Kristopher Attwood, Ram Nambiar, Jason B. Muhitch, Hanna R. Rosenheck, Renuka Iyer, Sarbajit Mukherjee, Christos Fountzilas, Pawel Kalinski, Agnieszka K. Witkiewicz, and Jennifer A. Jurcevic
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Cetuximab ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,Article ,Proto-Oncogene Proteins p21(ras) ,Immune system ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Microenvironment ,Cytotoxic T cell ,Humans ,Adverse effect ,Tumor microenvironment ,business.industry ,medicine.disease ,Primary tumor ,Fluorouracil ,business ,Colorectal Neoplasms ,medicine.drug - Abstract
Purpose: We evaluated the antitumor efficacy of cetuximab in combination with pembrolizumab in patients with RAS wild-type (RASwt), metastatic colorectal adenocarcinoma (mCRC). Patients and Methods: In this phase Ib/II study, cetuximab was combined with pembrolizumab in patients with RASwt mCRC with ≥ one prior line of therapy for advanced disease. We analyzed baseline on-treatment tumor tissues for changes in the tumor microenvironment (TME), using flow cytometry and multispectral immunofluorescence. Results: Forty-four patients were evaluable for efficacy. The study was negative for the primary efficacy endpoint [overall response rate: 2.6%, 6-month progression-free survival (PFS): 31%; P = 0.52]. Median PFS was 4.1 months [95% confidence interval (CI): 3.9–5.5 months]. No increase in adverse effects was identified. We observed favorable immunomodulation with 47% increase in the number of intratumoral CTLs posttreatment (P = 0.035). These changes were more pronounced in patients with tumor shrinkage (P = 0.05). The TME was characterized by high numbers of TIM3+ and CTLA4+ cells; there were few activated OX40+ cells. PD-L1 expression was higher in pretreatment tumor cells from metastatic sites versus primary tumor samples (P < 0.05). Higher numbers of PD-L1+ tumor cells at baseline were associated with tumor shrinkage (P = 0.04). Analysis of immune populations in the blood demonstrated decreases in PD-1+ memory effector cells (P = 0.04) and granulocytic myeloid-derived suppressor cells (P = 0.03), with simultaneous increases in CD4+/CTLA4+ cells (P = 0.01). Conclusions: The combination of cetuximab and pembrolizumab is inactive in patients with RASwt mCRC, despite its partial local immunologic efficacy. Further development of immuno-oncology combinations with enhanced efficacy and/or targeting additional or alternative immune checkpoints merits investigation.
- Published
- 2021
14. RB loss determines selective resistance and novel vulnerabilities in ER-positive breast cancer models
- Author
-
Vishnu Kumarasamy, Ram Nambiar, Jianxin Wang, Hanna Rosenheck, Agnieszka K. Witkiewicz, and Erik S. Knudsen
- Subjects
Cancer Research ,Cyclin-Dependent Kinase 4 ,Breast Neoplasms ,Cyclin-Dependent Kinase 6 ,Cytostatic Agents ,Retinoblastoma Protein ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Genetics ,Humans ,Cyclin D1 ,Female ,Molecular Biology - Abstract
The management of metastatic estrogen receptor (ER) positive HER2 negative breast cancer (ER+) has improved; however, therapeutic resistance and disease progression emerges in majority of cases. Using unbiased approaches, as expected PI3K and MTOR inhibitors emerge as potent inhibitors to delay proliferation of ER+ models harboring PIK3CA mutations. However, the cytostatic efficacy of these drugs is hindered due to marginal impact on the expression of cyclin D1. Different combination approaches involving the inhibition of ER pathway or cell cycle result in durable growth arrest via RB activation and subsequent inhibition of CDK2 activity. However, cell cycle alterations due to RB loss or ectopic CDK4/cyclin D1 activation yields resistance to these cytostatic combination treatments. To define means to counter resistance to targeted therapies imparted with RB loss; complementary drug screens were performed with RB-deleted isogenic cell lines. In this setting, RB loss renders ER+ breast cancer models more vulnerable to drugs that target DNA replication and mitosis. Pairwise combinations using these classes of drugs defines greater selectivity for RB deficiency. The combination of AURK and WEE1 inhibitors, yields synergistic cell death selectively in RB-deleted ER+ breast cancer cells via apoptosis and yields profound disease control in vivo. Through unbiased efforts the XIAP/CIAP inhibitor birinapant was identified as a novel RB-selective agent. Birinapant further enhances the cytotoxic effect of chemotherapies and targeted therapies used in the treatment of ER+ breast cancer models selectively in the RB-deficient setting. Using organoid culture and xenograft models, we demonstrate the highly selective use of birinapant based combinations for the treatment of RB-deficient tumors. Together, these data illustrate the critical role of RB-pathway in response to many agents used to treat ER+ breast cancer, whilst informing new therapeutic approaches that could be deployed against resistant disease.
- Published
- 2021
15. CDK/cyclin dependencies define extreme cancer cell-cycle heterogeneity and collateral vulnerabilities
- Author
-
Erik S. Knudsen, Vishnu Kumarasamy, Ram Nambiar, Joel D. Pearson, Paris Vail, Hanna Rosenheck, Jianxin Wang, Kevin Eng, Rod Bremner, Daniel Schramek, Seth M. Rubin, Alana L. Welm, and Agnieszka K. Witkiewicz
- Subjects
Neoplasms ,Cell Cycle ,Cyclin-Dependent Kinase 4 ,Humans ,Cell Cycle Proteins ,Cell Division ,Cyclin-Dependent Kinase Inhibitor p27 ,Cyclin-Dependent Kinases ,General Biochemistry, Genetics and Molecular Biology - Abstract
Progression through G1/S phase of the cell cycle is coordinated by cyclin-dependent kinase (CDK) activities. Here, we find that the requirement for different CDK activities and cyclins in driving cancer cell cycles is highly heterogeneous. The differential gene requirements associate with tumor origin and genetic alterations. We define multiple mechanisms for G1/S progression in RB-proficient models, which are CDK4/6 independent and elicit resistance to FDA-approved inhibitors. Conversely, RB-deficient models are intrinsically CDK4/6 independent, but exhibit differential requirements for cyclin E. These dependencies for CDK and cyclins associate with gene expression programs that denote intrinsically different cell-cycle states. Mining therapeutic sensitivities shows that there are reciprocal vulnerabilities associated with RB1 or CCND1 expression versus CCNE1 or CDKN2A. Together, these findings illustrate the complex nature of cancer cell cycles and the relevance for precision therapeutic intervention.
- Published
- 2022
16. Functional Determinants of Cell Cycle Plasticity and Sensitivity to CDK4/6 Inhibition
- Author
-
Ram Nambiar, Erik S. Knudsen, Vishnu Kumarasamy, Agnieszka K. Witkiewicz, and Paris Vail
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Pyridines ,Aminopyridines ,Context (language use) ,Antineoplastic Agents ,Breast Neoplasms ,Mice, Inbred Strains ,Palbociclib ,Piperazines ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Animals ,Humans ,Protein Kinase Inhibitors ,Cell Proliferation ,biology ,Kinase ,Chemistry ,MEK inhibitor ,Cyclin-dependent kinase 2 ,Cell Cycle ,Cyclin-Dependent Kinase 2 ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,Cell cycle ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Benzimidazoles ,CDK4/6 Inhibition - Abstract
Intrinsic or acquired resistance to clinically approved CDK4/6 inhibitors has emerged as a major obstacle that hinders their utility beyond ER+ breast cancer. In this study, CDK4/6-dependent and -resistant models were employed to identify functional determinants of response to pharmacologic CDK4/6 inhibitors. In all models tested, the activation of RB and inhibition of CDK2 activity emerged as determinants of sensitivity. While depleting CDK4 and 6 was sufficient to limit proliferation in specific resistance settings, RB loss rendered cells completely independent of these kinases. The main downstream target in this context was the activation status of CDK2, which was suppressed with CDK4/6 inhibition in an RB-dependent fashion. Protein levels of p27 were associated with plasticity/rigidity of the cell cycle and correlated with sensitivity to CDK4/6 inhibition. Exogenous overexpression and pharmacologic induction of p27 via inhibition of SKP2 and targeting the MEK/ERK pathway enhanced the cytostatic effect of CDK4/6 inhibitors. Mice bearing ER+ xenografts displayed a durable antitumor response to palbociclib; however, over the course of treatment, few cells retained RB phosphorylation, which was associated with limited p27 protein levels as determined by multispectral imaging. Similarly, combination treatment of palbociclib with a MEK inhibitor in pancreatic cancer PDX models upregulated p27 and further enhanced the in vivo tumor response to palbociclib. Collectively, these results suggest that the cell cycle plasticity, which enables tumor models to evade palbociclib-mediated activation of RB, could be targeted using a clinically applicable CDK2 inhibitor. Significance: This work provides a mechanistic insight toward understanding the functional roles of multiple cell cycle regulators that drive plasticity and sensitivity to CDK4/6 inhibition.
- Published
- 2020
17. Targeting dual signalling pathways in concert with immune checkpoints for the treatment of pancreatic cancer
- Author
-
Sejin Chung, Mukund Seshadri, Erik S. Knudsen, Vishnu Kumarasamy, Amanda Ruiz, Scott I. Abrams, Jin Wu, Stephanie L. Tzetzo, Shailender S. Chauhan, Paris Vail, Agnieszka K. Witkiewicz, Jared Sivinski, Jianmin Wang, and Ram Nambiar
- Subjects
0301 basic medicine ,Myeloid ,medicine.medical_treatment ,Antigen presentation ,Cell Culture Techniques ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Interferon ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Immune Checkpoint Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,business.industry ,Gastroenterology ,Cyclin-Dependent Kinase 4 ,Immunotherapy ,Cell Cycle Checkpoints ,Cyclin-Dependent Kinase 6 ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Pancreatic Neoplasms ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,business ,medicine.drug ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
ObjectiveThis study exploits the intersection between molecular-targeted therapies and immune-checkpoint inhibition to define new means to treat pancreatic cancer.DesignPatient-derived cell lines and xenograft models were used to define the response to CDK4/6 and MEK inhibition in the tumour compartment. Impacts relative to immunotherapy were performed using subcutaneous and orthotopic syngeneic models. Single-cell RNA sequencing and multispectral imaging were employed to delineate effects on the immunological milieu in the tumour microenvironment.ResultsWe found that combination treatment with MEK and CDK4/6 inhibitors was effective across a broad range of PDX models in delaying tumour progression. These effects were associated with stable cell-cycle arrest, as well as the induction of multiple genes associated with interferon response and antigen presentation in an RB-dependent fashion. Using single-cell sequencing and complementary approaches, we found that the combination of CDK4/6 and MEK inhibition had a significant impact on increasing T-cell infiltration and altering myeloid populations, while potently cooperating with immune checkpoint inhibitors.ConclusionsTogether, these data indicate that there are canonical and non-canonical features of CDK4/6 and MEK inhibition that impact on the tumour and immune microenvironment. This combination-targeted treatment can promote robust tumour control in combination with immune checkpoint inhibitor therapy.
- Published
- 2020
18. PolyA_DB 3 catalogs cleavage and polyadenylation sites identified by deep sequencing in multiple genomes
- Author
-
Ruijia Wang, Ram Nambiar, Dinghai Zheng, and Bin Tian
- Subjects
0301 basic medicine ,Polyadenylation ,Sequence analysis ,Computational biology ,Genome browser ,Biology ,Cleavage (embryo) ,Genome ,Deep sequencing ,Mice ,User-Computer Interface ,03 medical and health sciences ,0302 clinical medicine ,Databases, Genetic ,Genetics ,Database Issue ,Animals ,Humans ,Gene ,RNA Cleavage ,Expressed sequence tag ,Sequence Analysis, RNA ,High-Throughput Nucleotide Sequencing ,Rats ,030104 developmental biology ,Chickens ,030217 neurology & neurosurgery - Abstract
PolyA_DB is a database cataloging cleavage and polyadenylation sites (PASs) in several genomes. Previous versions were based mainly on expressed sequence tags (ESTs), which had a limited amount and could lead to inaccurate PAS identification due to the presence of internal A-rich sequences in transcripts. Here, we present an updated version of the database based solely on deep sequencing data. First, PASs are mapped by the 3′ region extraction and deep sequencing (3′READS) method, ensuring unequivocal PAS identification. Second, a large volume of data based on diverse biological samples increases PAS coverage by 3.5-fold over the EST-based version and provides PAS usage information. Third, strand-specific RNA-seq data are used to extend annotated 3′ ends of genes to obtain more thorough annotations of alternative polyadenylation (APA) sites. Fourth, conservation information of PAS across mammals sheds light on significance of APA sites. The database (URL: http://www.polya-db.org/v3) currently holds PASs in human, mouse, rat and chicken, and has links to the UCSC genome browser for further visualization and for integration with other genomic data.
- Published
- 2017
19. Transmission of a newly characterized strain of varicella-zoster virus from a patient with herpes zoster in a long-term-care facility, West Virginia, 2004
- Author
-
Ram Nambiar, Andrea N. Burnett-Hartman, D. Scott Schmid, Dalya Guris, Linda Ritz, Patricia Owens, Vladimir N. Loparev, and Adriana S. Lopez
- Subjects
Adult ,Male ,Herpesvirus 3, Human ,Infectious Disease Transmission, Patient-to-Professional ,Genotype ,viruses ,medicine.disease_cause ,Antiviral Agents ,Herpes Zoster ,Herpesviridae ,Virus ,Article ,Disease Outbreaks ,Chickenpox Vaccine ,Chickenpox ,Alphaherpesvirinae ,Immunology and Allergy ,Medicine ,Humans ,Phylogeny ,Aged, 80 and over ,Cross Infection ,biology ,integumentary system ,business.industry ,Varicella zoster virus ,Outbreak ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Middle Aged ,West Virginia ,medicine.disease ,biology.organism_classification ,Virology ,Long-Term Care ,eye diseases ,Infectious Diseases ,Fomites ,Immunology ,DNA, Viral ,Female ,Viral disease ,business - Abstract
We investigated a small outbreak of varicella in a long-term-care facility after a case of herpes zoster. Clinical specimens and environmental samples were collected from all case patients and from surfaces in the case patients' rooms and other common-use areas. Wild-type varicella-zoster virus (VZV) DNA was identified in all 3 varicella case patients, and high concentrations of VZV DNA were detected in environmental samples from the room of the herpes zoster case patient. Genotypic analysis showed that the identical VZV strain was present in all samples; moreover, the strain was a unique Mosaic genotype isolate that included a stable Oka vaccine marker that had hitherto never been observed in a wild-type strain of VZV. This study provides evidence for the value of including environmental sampling during the investigation of varicella outbreaks and illustrates the importance of evaluating multiple vaccine-associated markers for the discrimination of vaccine virus from wild-type VZV.
- Published
- 2008
20. A cytological study of natural hybrids between Prosimulium multidentatum and P. magnum
- Author
-
Klaus Rothfels and Ram Nambiar
- Subjects
Genetics ,Sympatry ,Meiosis ,Backcrossing ,Centromere ,Chromosome ,Zoology ,Biology ,Chromosome pairing ,Genetics (clinical) ,Chiasma ,Hybrid - Abstract
In a limited area of sympatry in New York State, hybridization occurs between Prosimulium multidentatum (Twinn) and “forms” 2 and 3 of P. magnum Dyar and Shannon. Salivary gland chromosomes of the interspecific hybrids are the composite of those of the parental species with respect to fixed inversion differences, sex chromosomes and chromocenter attachment of centromeres. Backcross hybrids with P. multidentatum occur in the same area. The preferred direction of cross (P. multidentatum ♀ x P. magnum ♂) is rationalized by the earlier emergence of P. multidentatum, and the earlier emergence of males and greater longevity of females of both species. Chromosome pairing, both in F1 and backcross, is loose in hybrid segments and tight in species homozygous parts. Likewise chiasma frequency is low in hybrid as compared to parentally homozygous chromosomes. In the face of locally recurrent hybridization species integrity is maintained by meiotic irregularities in the hybrids and especially by disturbances in the sex chromosome balance of backcross hybrids, the sex chromosomes of P. multidentatum and P. magnum being non-homologous.
- Published
- 1981
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.