247 results on '"Duncan I. Jodrell"'
Search Results
2. Quantifying cell cycle-dependent drug sensitivities in cancer using a high throughput synchronisation and screening approach
- Author
-
Timothy I. Johnson, Christopher J. Minteer, Daniel Kottmann, Charles R. Dunlop, Sandra Bernaldo de Quirós Fernández, Larissa S. Carnevalli, Yann Wallez, Alan Lau, Frances M. Richards, and Duncan I. Jodrell
- Subjects
Cancer ,cell cycle ,synchronisation ,high throughput screening ,combination treatment ,Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Chemotherapy and targeted agent anti-cancer efficacy is largely dependent on the proliferative state of tumours, as exemplified by agents that target DNA synthesis/replication or mitosis. As a result, cell cycle specificities of a number of cancer drugs are well known. However, they are yet to be described in a quantifiable manner. Methods: A scalable cell synchronisation protocol used to screen a library of 235 anti-cancer compounds exposed over six hours in G1 or S/G2 accumulated AsPC-1 cells to generate a cell cycle specificity (CCS) score. Findings: The synchronisation method was associated with reduced method-related cytotoxicity compared to nocodazole, delivering sufficient cell cycle purity and cell numbers to run high-throughput drug library screens. Compounds were identified with G1 and S/G2-associated specificities that, overall, functionally matched with a compound's target/mechanism of action. This annotation was used to describe a synergistic schedule using the CDK4/6 inhibitor, palbociclib, prior to gemcitabine/AZD6738 as well as describe the correlation between the CCS score and published synergistic/antagonistic drug schedules. Interpretation: This is the first highly quantitative description of cell cycle-dependent drug sensitivities that utilised a tractable and tolerated method with potential uses outside the present study. Drug treatments such as those shown to be G1 or S/G2 associated may benefit from scheduling considerations such as after CDK4/6 inhibitors and being first in drug sequences respectively. Funding: Cancer Research UK (CRUK) Institute core grants C14303/A17197 and C9545/A29580. The Li Ka Shing Centre where this work was performed was generously funded by CK Hutchison Holdings Limited, the University of Cambridge, CRUK, The Atlantic Philanthropies and others.
- Published
- 2021
- Full Text
- View/download PDF
3. Targeting melanoma’s MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors
- Author
-
Matthew J. Sale, Emma Minihane, Noel R. Monks, Rebecca Gilley, Frances M. Richards, Kevin P. Schifferli, Courtney L. Andersen, Emma J. Davies, Mario Aladren Vicente, Eiko Ozono, Aleksandra Markovets, Jonathan R. Dry, Lisa Drew, Vikki Flemington, Theresa Proia, Duncan I. Jodrell, Paul D. Smith, and Simon J. Cook
- Subjects
Science - Abstract
BRAF or MEK1/2 inhibitors are cytostatic in melanoma and the surviving cells develop drug resistance. This study shows that the pro-survival pool is biased towards MCL1 in melanoma so that BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, improving tumour growth inhibition.
- Published
- 2019
- Full Text
- View/download PDF
4. Quantifying Drug‐Induced Bone Marrow Toxicity Using a Novel Haematopoiesis Systems Pharmacology Model
- Author
-
Chiara Fornari, Lenka Oplustil O'Connor, Carmen Pin, Aaron Smith, James W.T. Yates, S.Y. Amy Cheung, Duncan I. Jodrell, Jerome T. Mettetal, and Teresa A. Collins
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Haematological toxicity associated with cancer therapeutics is monitored by changes in blood cell count, and their primary effect is on proliferative progenitors in the bone marrow. Using observations in rat bone marrow and blood, we characterize a mathematical model that comprises cell proliferation and differentiation of the full haematopoietic phylogeny, with interacting feedback loops between lineages in homeostasis as well as following carboplatin exposure. We accurately predicted the temporal dynamics of several mature cell types related to carboplatin‐induced bone marrow toxicity and identified novel insights into haematopoiesis. Our model confirms a significant degree of plasticity within bone marrow cells, with the number and type of both early progenitors and circulating cells affecting cell balance, via feedback mechanisms, through fate decisions of the multipotent progenitors. We also demonstrated cross‐species translation of our predictions to patients, applying the same core model structure and considering differences in drug‐dependent and physiology‐dependent parameters.
- Published
- 2019
- Full Text
- View/download PDF
5. SPARC dependent collagen deposition and gemcitabine delivery in a genetically engineered mouse model of pancreas cancerResearch in context
- Author
-
Iswarya Ramu, Sören M. Buchholz, Melanie S. Patzak, Robert G. Goetze, Shiv K. Singh, Frances M. Richards, Duncan I. Jodrell, Bence Sipos, Philipp Ströbel, Volker Ellenrieder, Elisabeth Hessmann, and Albrecht Neesse
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is characterised by extensive matrix deposition that has been implicated in impaired drug delivery and therapeutic resistance. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that regulates collagen deposition and is highly upregulated in the activated stroma subtype with poor prognosis in PDAC patients. Methods: KrasG12D;p48-Cre;SPARC−/− (KC-SPARC−/−) and KrasG12D;p48-Cre;SPARCWT (KC-SPARCWT) were generated and analysed at different stages of carcinogenesis by histological grading, immunohistochemistry for epithelial and stromal markers, survival and preclinical analysis. Pharmacokinetic and pharmacodynamic studies were conducted by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and immunohistochemistry following gemcitabine treatment (100 mg/kg) in vivo. Findings: Global genetic ablation of SPARC in a KrasG12D driven mouse model resulted in significantly reduced overall and mature collagen deposition around early and advanced pancreatic intraepithelial neoplasia (PanIN) lesions and in invasive PDAC (p
- Published
- 2019
- Full Text
- View/download PDF
6. Cytosolic 5′-nucleotidase 1A is overexpressed in pancreatic cancer and mediates gemcitabine resistance by reducing intracellular gemcitabine metabolitesResearch in context
- Author
-
Melanie S. Patzak, Vijayalakshmi Kari, Shilpa Patil, Feda H. Hamdan, Robert G. Goetze, Marius Brunner, Jochen Gaedcke, Julia Kitz, Duncan I. Jodrell, Frances M. Richards, Christian Pilarsky, Robert Gruetzmann, Petra Rümmele, Thomas Knösel, Elisabeth Hessmann, Volker Ellenrieder, Steven A. Johnsen, and Albrecht Neesse
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Cytosolic 5′-nucleotidase 1A (NT5C1A) dephosphorylates non-cyclic nucleoside monophosphates to produce nucleosides and inorganic phosphates. Here, we investigate NT5C1A expression in pancreatic ductal adenocarcinoma (PDAC) and its impact on gemcitabine metabolism and therapeutic efficacy. Methods: NT5C1A expression was determined by semiquantitative immunohistochemistry using tissue microarrays. Gemcitabine metabolites and response were assessed in several human and murine PDAC cell lines using crystal violet assays, Western blot, viability assays, and liquid chromatography tandem mass-spectrometry (LC-MS/MS). Findings: NT5C1A was strongly expressed in tumor cells of a large subgroup of resected PDAC patients in two independent patient cohorts (44–56% score 2 and 8–26% score 3, n = 414). In contrast, NT5C1A was expressed at very low levels in the tumor stroma, and neither stromal nor tumoral expression was a prognostic marker for postoperative survival. In vitro, NT5C1A overexpression increased gemcitabine resistance by reducing apoptosis levels and significantly decreased intracellular amounts of cytotoxic dFdCTP in +NT5C1A tumor cells. Co-culture experiments with conditioned media from +NT5C1A PSCs improved gemcitabine efficacy in tumor cells. In vivo, therapeutic efficacy of gemcitabine was significantly decreased and serum levels of the inactive gemcitabine metabolite dFdU significantly increased in mice bearing NT5C1A overexpressing tumors. Interpretation: NT5C1A is robustly expressed in tumor cells of resected PDAC patients. Moreover, NT5C1A mediates gemcitabine resistance by decreasing the amount of intracellular dFdCTP, leading to reduced tumor cell apoptosis and larger pancreatic tumors in mice. Further studies should clarify the role of NT5C1A as novel predictor for gemcitabine treatment response in patients with PDAC. Keywords: Cytosolic 5′-nucleotidase 1A, Chemotherapeutic resistance, Gemcitabine, Pancreatic cancer
- Published
- 2019
- Full Text
- View/download PDF
7. CSF1R+ Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype
- Author
-
Juliana B. Candido, Jennifer P. Morton, Peter Bailey, Andrew D. Campbell, Saadia A. Karim, Thomas Jamieson, Laura Lapienyte, Aarthi Gopinathan, William Clark, Ewan J. McGhee, Jun Wang, Monica Escorcio-Correia, Raphael Zollinger, Rozita Roshani, Lisa Drew, Loveena Rishi, Rebecca Arkell, T.R. Jeffry Evans, Colin Nixon, Duncan I. Jodrell, Robert W. Wilkinson, Andrew V. Biankin, Simon T. Barry, Frances R. Balkwill, and Owen J. Sansom
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Pancreatic ductal adenocarcinoma (PDAC) is resistant to most therapies including single-agent immunotherapy and has a dense desmoplastic stroma, and most patients present with advanced metastatic disease. We reveal that macrophages are the dominant leukocyte population both in human PDAC stroma and autochthonous models, with an important functional contribution to the squamous subtype of human PDAC. We targeted macrophages in a genetic PDAC model using AZD7507, a potent selective inhibitor of CSF1R. AZD7507 caused shrinkage of established tumors and increased mouse survival in this difficult-to-treat model. Malignant cell proliferation diminished, with increased cell death and an enhanced T cell immune response. Loss of macrophages rewired other features of the TME, with global changes in gene expression akin to switching PDAC subtypes. These changes were markedly different to those elicited when neutrophils were targeted via CXCR2. These results suggest targeting the myeloid cell axis may be particularly efficacious in PDAC, especially with CSF1R inhibitors. : Candido et al. find that CSF-1R+ macrophages play a vital role in pancreatic tumor maintenance, suppression of T cells, and tumor gene networks based on transcriptome analysis. In an autochthonous model, CSF-1R inhibition alters the gene expression programs that influence subtype specification of PDAC and extends survival. Keywords: pancreatic cancer, macrophages, CSF1R, T cells
- Published
- 2018
- Full Text
- View/download PDF
8. GEMMs as preclinical models for testing pancreatic cancer therapies
- Author
-
Aarthi Gopinathan, Jennifer P. Morton, Duncan I. Jodrell, and Owen J. Sansom
- Subjects
Co-clinical trials ,Preclinical mouse models ,Pancreatic ductal adenocarcinoma ,PDAC ,Drug discovery ,Drug development ,Medicine ,Pathology ,RB1-214 - Abstract
Pancreatic ductal adenocarcinoma is the most common form of pancreatic tumour, with a very limited survival rate and currently no available disease-modifying treatments. Despite recent advances in the production of genetically engineered mouse models (GEMMs), the development of new therapies for pancreatic cancer is still hampered by a lack of reliable and predictive preclinical animal models for this disease. Preclinical models are vitally important for assessing therapies in the first stages of the drug development pipeline, prior to their transition to the clinical arena. GEMMs carry mutations in genes that are associated with specific human diseases and they can thus accurately mimic the genetic, phenotypic and physiological aspects of human pathologies. Here, we discuss different GEMMs of human pancreatic cancer, with a focus on the Lox-Stop-Lox (LSL)-KrasG12D; LSL-Trp53R172H; Pdx1-cre (KPC) model, one of the most widely used preclinical models for this disease. We describe its application in preclinical research, highlighting its advantages and disadvantages, its potential for predicting clinical outcomes in humans and the factors that can affect such outcomes, and, finally, future developments that could advance the discovery of new therapies for pancreatic cancer.
- Published
- 2015
- Full Text
- View/download PDF
9. Supplementary Table 1, Figures 1-6 from nab-Paclitaxel Potentiates Gemcitabine Activity by Reducing Cytidine Deaminase Levels in a Mouse Model of Pancreatic Cancer
- Author
-
David A. Tuveson, Duncan I. Jodrell, Martijn P. Lolkema, Tashinga E. Bapiro, Natalie Cook, Albrecht Neesse, and Kristopher K. Frese
- Abstract
PDF file - 392K
- Published
- 2023
10. Figure S2 from The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression
- Author
-
Duncan I. Jodrell, Frances M. Richards, Alan Lau, Sandra Bernaldo de Quirós Fernández, James W.T. Yates, Chiara Fornari, Siang-Boon Koh, Timothy Isaac Johnson, Charles R. Dunlop, and Yann Wallez
- Abstract
AZD6738 and gemcitabine synergistically inhibit cell growth in a panel of PDAC cell lines
- Published
- 2023
11. Interview with Dr. Tuveson from nab-Paclitaxel Potentiates Gemcitabine Activity by Reducing Cytidine Deaminase Levels in a Mouse Model of Pancreatic Cancer
- Author
-
David A. Tuveson, Duncan I. Jodrell, Martijn P. Lolkema, Tashinga E. Bapiro, Natalie Cook, Albrecht Neesse, and Kristopher K. Frese
- Abstract
mp3 file (10 MB). In the March edition of the Cancer Discovery podcast, Executive Editor Mark Landis talks with David A. Tuveson about his paper, which provides mechanistic insight into the clinical cooperation observed between gemcitabine and nab-paclitaxel in the treatment of pancreatic cancer.
- Published
- 2023
12. Data from The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression
- Author
-
Duncan I. Jodrell, Frances M. Richards, Alan Lau, Sandra Bernaldo de Quirós Fernández, James W.T. Yates, Chiara Fornari, Siang-Boon Koh, Timothy Isaac Johnson, Charles R. Dunlop, and Yann Wallez
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers, and overall survival rates have barely improved over the past five decades. The antimetabolite gemcitabine remains part of the standard of care but shows very limited antitumor efficacy. Ataxia telangiectasia and Rad3-related protein (ATR), the apical kinase of the intra–S-phase DNA damage response, plays a central role in safeguarding cells from replication stress and can therefore limit the efficacy of antimetabolite drug therapies. We investigated the ability of the ATR inhibitor, AZD6738, to prevent the gemcitabine-induced intra–S-phase checkpoint activation and evaluated the antitumor potential of this combination in vitro and in vivo. In PDAC cell lines, AZD6738 inhibited gemcitabine-induced Chk1 activation, prevented cell-cycle arrest, and restrained RRM2 accumulation, leading to the strong induction of replication stress markers only with the combination. Moreover, synergistic growth inhibition was identified in a panel of 5 mouse and 7 human PDAC cell lines using both Bliss Independence and Loewe models. In clonogenic assays, the combination abrogated survival at concentrations for which single agents had minor effects. In vivo, AZD6738 in combination with gemcitabine was well tolerated and induced tumor regression in a subcutaneous allograft model of a KrasG12D; Trp53R172H; Pdx-Cre (KPC) mouse cancer cell line, significantly extending survival. Remarkably, the combination also induced regression of a subgroup of KPC autochthonous tumors, which generally do not respond well to conventional chemotherapy. Altogether, our data suggest that AZD6738 in combination with gemcitabine merits evaluation in a clinical trial in patients with PDAC. Mol Cancer Ther; 17(8); 1670–82. ©2018 AACR.
- Published
- 2023
13. Data from nab-Paclitaxel Potentiates Gemcitabine Activity by Reducing Cytidine Deaminase Levels in a Mouse Model of Pancreatic Cancer
- Author
-
David A. Tuveson, Duncan I. Jodrell, Martijn P. Lolkema, Tashinga E. Bapiro, Natalie Cook, Albrecht Neesse, and Kristopher K. Frese
- Abstract
Nanoparticle albumin-bound (nab)-paclitaxel, an albumin-stabilized paclitaxel formulation, demonstrates clinical activity when administered in combination with gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma (PDA). The limited availability of patient tissue and exquisite sensitivity of xenografts to chemotherapeutics have limited our ability to address the mechanistic basis of this treatment regimen. Here, we used a mouse model of PDA to show that the coadministration of nab-paclitaxel and gemcitabine uniquely demonstrates evidence of tumor regression. Combination treatment increases intratumoral gemcitabine levels attributable to a marked decrease in the primary gemcitabine metabolizing enzyme, cytidine deaminase. Correspondingly, paclitaxel reduced the levels of cytidine deaminase protein in cultured cells through reactive oxygen species–mediated degradation, resulting in the increased stabilization of gemcitabine. Our findings support the concept that suboptimal intratumoral concentrations of gemcitabine represent a crucial mechanism of therapeutic resistance in PDA and highlight the advantages of genetically engineered mouse models in preclinical therapeutic trials.Significance: This study provides mechanistic insight into the clinical cooperation observed between gemcitabine and nab-paclitaxel in the treatment of pancreatic cancer. Cancer Discovery; 2(3); 260–9. ©2012 AACR.Read the Commentary on this article by Clark, p. 208This article is highlighted in the In This Issue feature, p. 193
- Published
- 2023
14. Table S1 from The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression
- Author
-
Duncan I. Jodrell, Frances M. Richards, Alan Lau, Sandra Bernaldo de Quirós Fernández, James W.T. Yates, Chiara Fornari, Siang-Boon Koh, Timothy Isaac Johnson, Charles R. Dunlop, and Yann Wallez
- Abstract
Genetic alterations likely to sensitize to ATR inhibition in the human PDAC cell lines
- Published
- 2023
15. Supplementary Methods, Figure Legends 1-6, Table 1 from nab-Paclitaxel Potentiates Gemcitabine Activity by Reducing Cytidine Deaminase Levels in a Mouse Model of Pancreatic Cancer
- Author
-
David A. Tuveson, Duncan I. Jodrell, Martijn P. Lolkema, Tashinga E. Bapiro, Natalie Cook, Albrecht Neesse, and Kristopher K. Frese
- Abstract
PDF file - 122K
- Published
- 2023
16. Supplementary Data from Phase I Safety, Pharmacokinetics, and Inhibition of Src Activity Study of Saracatinib in Patients with Solid Tumors
- Author
-
Josep Tabernero, Renee Iacona, Alan Swaisland, Paul Elvin, Esther Casado, Paul Hamberg, Duncan I. Jodrell, Herbert I. Hurwitz, Klaas Hoekman, Isabel Chirivella, Erika Martinelli, Andres Cervantes, and José Baselga
- Abstract
Supplementary Figure S1; Supplementary Appendix.
- Published
- 2023
17. Data from Phase I Safety, Pharmacokinetics, and Inhibition of Src Activity Study of Saracatinib in Patients with Solid Tumors
- Author
-
Josep Tabernero, Renee Iacona, Alan Swaisland, Paul Elvin, Esther Casado, Paul Hamberg, Duncan I. Jodrell, Herbert I. Hurwitz, Klaas Hoekman, Isabel Chirivella, Erika Martinelli, Andres Cervantes, and José Baselga
- Abstract
Purpose: This dose-escalation study evaluated the safety, tolerability, and pharmacokinetics (PK) of the oral Src inhibitor saracatinib (AZD0530) in patients with advanced solid malignancies. Tumor biopsy samples were taken to investigate the effect of saracatinib on Src activity in tumors.Experimental Design: Part A of the study followed a multiple-ascending dose design to establish the maximum tolerated dose (MTD) of saracatinib. Part B was a randomized, parallel-group, cohort-expansion phase to further assess tolerated doses. Safety, tolerability, and Src activity (immunohistochemistry and lysate-based methodologies) were assessed after 21 days of once-daily oral dosing. PK was assessed after single and multiple dosing.Results: In part A, 30 patients received once-daily saracatinib at doses of 60 to 250 mg; the MTD was established as 175 mg. In part B, 51 patients were randomized to receive 50 mg (n = 16), 125 mg (n = 16), or 175 mg (n = 19) of saracatinib. The most common grade ≥3 events considered to be treatment related were anemia, diarrhea, and asthenia. Tumor Src activity was reduced following saracatinib treatment. The area under the concentration-time curve and Cmax of saracatinib increased with increasing dose. Saracatinib accumulated 4- to 5-fold on once-daily dosing to reach steady-state exposure after 10 to 17 days of dosing. The half-life was ∼40 hours.Conclusions: Saracatinib was well tolerated in patients with advanced solid malignancies. A reduction in tumor Src activity was observed. PK data show that saracatinib is suitable for once-daily oral dosing. Based on this study, the recommended dose for the phase II studies was chosen to be 175 mg/d. Clin Cancer Res; 16(19); 4876–83. ©2010 AACR.
- Published
- 2023
18. Supplemental Methods from mTORC2 Signaling Drives the Development and Progression of Pancreatic Cancer
- Author
-
Jennifer P. Morton, Brian C. Lewis, Owen J. Sansom, Simon T. Barry, David S. Klimstra, Brian J. Quattrochi, Michael N. Hall, Nabeel Bardeesy, T.R. Jeffry Evans, Duncan I. Jodrell, Aarthi Gopinathan, Yusuke Mizukami, Jun Yu, Wright Jacob, David M. Gay, Makoto Sano, Saadia A Karim, and David R. Driscoll
- Abstract
Supplementary materials and methods
- Published
- 2023
19. Data from Identification of Resistance Pathways Specific to Malignancy Using Organoid Models of Pancreatic Cancer
- Author
-
David A. Tuveson, Youngkyu Park, Duncan I. Jodrell, Kenneth H. Yu, Paul Smith, Pearl Huang, Tashinga E. Bapiro, Kevin Wright, Dea Filippini, Lindsey A. Baker, Iok In Christine Chio, Daniel Öhlund, Chang-Il Hwang, Tobiloba E. Oni, Kristopher K. Frese, Dannielle D. Engle, Hervé Tiriac, Vincenzo Corbo, and Mariano Ponz-Sarvise
- Abstract
Purpose:KRAS is mutated in the majority of pancreatic ductal adenocarcinoma. MAPK and PI3K-AKT are primary KRAS effector pathways, but combined MAPK and PI3K inhibition has not been demonstrated to be clinically effective to date. We explore the resistance mechanisms uniquely employed by malignant cells.Experimental Design:We evaluated the expression and activation of receptor tyrosine kinases in response to combined MEK and AKT inhibition in KPC mice and pancreatic ductal organoids. In addition, we sought to determine the therapeutic efficacy of targeting resistance pathways induced by MEK and AKT inhibition in order to identify malignant-specific vulnerabilities.Results:Combined MEK and AKT inhibition modestly extended the survival of KPC mice and increased Egfr and ErbB2 phosphorylation levels. Tumor organoids, but not their normal counterparts, exhibited elevated phosphorylation of ERBB2 and ERBB3 after MEK and AKT blockade. A pan-ERBB inhibitor synergized with MEK and AKT blockade in human PDA organoids, whereas this was not observed for the EGFR inhibitor erlotinib. Combined MEK and ERBB inhibitor treatment of human organoid orthotopic xenografts was sufficient to cause tumor regression in short-term intervention studies.Conclusions:Analyses of normal and tumor pancreatic organoids revealed the importance of ERBB activation during MEK and AKT blockade primarily in the malignant cultures. The lack of ERBB hyperactivation in normal organoids suggests a larger therapeutic index. In our models, pan-ERBB inhibition was synergistic with dual inhibition of MEK and AKT, and the combination of a pan-ERBB inhibitor with MEK antagonists showed the highest activity both in vitro and in vivo.
- Published
- 2023
20. Supplemental Figure Legends from mTORC2 Signaling Drives the Development and Progression of Pancreatic Cancer
- Author
-
Jennifer P. Morton, Brian C. Lewis, Owen J. Sansom, Simon T. Barry, David S. Klimstra, Brian J. Quattrochi, Michael N. Hall, Nabeel Bardeesy, T.R. Jeffry Evans, Duncan I. Jodrell, Aarthi Gopinathan, Yusuke Mizukami, Jun Yu, Wright Jacob, David M. Gay, Makoto Sano, Saadia A Karim, and David R. Driscoll
- Abstract
Supplementary figure legends for Figures S1-S8
- Published
- 2023
21. Data from Targeting Carcinoembryonic Antigen with DNA Vaccination: On-Target Adverse Events Link with Immunologic and Clinical Outcomes
- Author
-
Christian H. Ottensmeier, Freda K. Stevenson, Gareth J. Thomas, Ugur Sahin, Petra Simon, Toni Weinschenk, Duncan I. Jodrell, Alan Anthoney, Sally Clive, Ann O'Callaghan, Andrew Bateman, Sarah Halford, Ceri Edwards, Emily Buxton, Paul Lloyd-Evans, Andrew King, Stephen M. Thirdborough, Jana Stasakova, Lindsey Chudley, Angelica Cazaly, Ann Mander, and Katy J. McCann
- Abstract
Purpose: We have clinically evaluated a DNA fusion vaccine to target the HLA-A*0201–binding peptide CAP-1 from carcinoembryonic antigen (CEA605–613) linked to an immunostimulatory domain (DOM) from fragment C of tetanus toxin.Experimental Design: Twenty-seven patients with CEA-expressing carcinomas were recruited: 15 patients with measurable disease (arm-I) and 12 patients without radiological evidence of disease (arm-II). Six intramuscular vaccinations of naked DNA (1 mg/dose) were administered up to week 12. Clinical and immunologic follow-up was up to week 64 or clinical/radiological disease.Results: DOM-specific immune responses demonstrated successful vaccine delivery. All patients without measurable disease compared with 60% with advanced disease responded immunologically, while 58% and 20% expanded anti-CAP-1 CD8+ T cells, respectively. CAP-1–specific T cells were only detectable in the blood postvaccination but could also be identified in previously resected cancer tissue. The gastrointestinal adverse event diarrhea was reported by 48% of patients and linked to more frequent decreases in CEA (P < 0.001) and improved global immunologic responses [anti-DOM responses of greater magnitude (P < 0.001), frequency (P = 0.004), and duration] compared with patients without diarrhea. In advanced disease patients, decreases in CEA were associated with better overall survival (HR = 0.14, P = 0.017). CAP-1 peptide was detectable on MHC class I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhea through CD8+ T-cell attack.Conclusions: Our data suggest that DNA vaccination is able to overcome peripheral tolerance in normal and tumor tissue and warrants testing in combination studies, for example, by vaccinating in parallel to treatment with an anti-PD1 antibody. Clin Cancer Res; 22(19); 4827–36. ©2016 AACR.
- Published
- 2023
22. Supplementary Data from Identification of Resistance Pathways Specific to Malignancy Using Organoid Models of Pancreatic Cancer
- Author
-
David A. Tuveson, Youngkyu Park, Duncan I. Jodrell, Kenneth H. Yu, Paul Smith, Pearl Huang, Tashinga E. Bapiro, Kevin Wright, Dea Filippini, Lindsey A. Baker, Iok In Christine Chio, Daniel Öhlund, Chang-Il Hwang, Tobiloba E. Oni, Kristopher K. Frese, Dannielle D. Engle, Hervé Tiriac, Vincenzo Corbo, and Mariano Ponz-Sarvise
- Abstract
Supplementary Figures 1-3
- Published
- 2023
23. Data from mTORC2 Signaling Drives the Development and Progression of Pancreatic Cancer
- Author
-
Jennifer P. Morton, Brian C. Lewis, Owen J. Sansom, Simon T. Barry, David S. Klimstra, Brian J. Quattrochi, Michael N. Hall, Nabeel Bardeesy, T.R. Jeffry Evans, Duncan I. Jodrell, Aarthi Gopinathan, Yusuke Mizukami, Jun Yu, Wright Jacob, David M. Gay, Makoto Sano, Saadia A Karim, and David R. Driscoll
- Abstract
mTOR signaling controls several critical cellular functions and is deregulated in many cancers, including pancreatic cancer. To date, most efforts have focused on inhibiting the mTORC1 complex. However, clinical trials of mTORC1 inhibitors in pancreatic cancer have failed, raising questions about this therapeutic approach. We employed a genetic approach to delete the obligate mTORC2 subunit Rictor and identified the critical times during which tumorigenesis requires mTORC2 signaling. Rictor deletion resulted in profoundly delayed tumorigenesis. Whereas previous studies showed most pancreatic tumors were insensitive to rapamycin, treatment with a dual mTORC1/2 inhibitor strongly suppressed tumorigenesis. In late-stage tumor-bearing mice, combined mTORC1/2 and PI3K inhibition significantly increased survival. Thus, targeting mTOR may be a potential therapeutic strategy in pancreatic cancer. Cancer Res; 76(23); 6911–23. ©2016 AACR.
- Published
- 2023
24. Supplementary Figures S1-S5 from CHK1 Inhibition Synergizes with Gemcitabine Initially by Destabilizing the DNA Replication Apparatus
- Author
-
Duncan I. Jodrell, Frances M. Richards, Robert G. Boyle, Richard J. Boyce, Aurélie Courtin, and Siang-Boon Koh
- Abstract
Supplementary Figures S1-S5. Effective synergy at sub-GI50 concentrations (S1); Mitotic entry at synergistic concentrations (S2); Cell fate upon gemcitabine and CHK1 inhibition (S3); Replication stress upon gemcitabine and CHK1 inhibition (S4); Mitotic aberration upon gemcitabine and CHK1 inhibition (S5).
- Published
- 2023
25. Supplementary Methods and References from CHK1 Inhibition Synergizes with Gemcitabine Initially by Destabilizing the DNA Replication Apparatus
- Author
-
Duncan I. Jodrell, Frances M. Richards, Robert G. Boyle, Richard J. Boyce, Aurélie Courtin, and Siang-Boon Koh
- Abstract
Supplementary Methods and References. Description of additional methods and procedures used in the study. Also includes Supplementary References.
- Published
- 2023
26. Supplementary Tables S1-S2 from CHK1 Inhibition Synergizes with Gemcitabine Initially by Destabilizing the DNA Replication Apparatus
- Author
-
Duncan I. Jodrell, Frances M. Richards, Robert G. Boyle, Richard J. Boyce, Aurélie Courtin, and Siang-Boon Koh
- Abstract
Supplementary Tables S1-S2. Kinase specificity of S1181 (S1); Percentages of S-phase cells (S2).
- Published
- 2023
27. Fate-mapping lymphocyte clones and their progenies from induced antigen-signals identifies temporospatial behaviours of T cells mediating tolerance
- Author
-
Munetomo Takahashi, Tsz Y. So, Kate Williamson, Zhaleh Hosseini, Katarzyna Kania, Michelle Ruhle, Tiffeney Mann, Martijn J. Schujis, Paul Coupland, Dean Naisbitt, Timotheus Y.F. Halim, Paul A. Lyons, Pietro Lio, Klaus Okkenhaug, David J. Adams, Ken G.C. Smith, Duncan I. Jodrell, Michael A. Chapman, and James E. D. Thaventhiran
- Abstract
Tissue homeostasis is maintained by the behaviours of lymphocyte clones responding to antigenic triggers in the face of pathogen, environmental, and developmental challenges. Current methodologies for tracking the behaviour of specific lymphocytes identify clones of a defined antigen-receptor—antigen binding affinity. However, lymphocytes can receive antigenic signals from undefined or endogenous antigens, and the strength of each signal, even for the same lymphocyte, varies with accessory signalling, across tissues and across time. We present a novel fate-mapping mouse, that, by tracking lymphocyte clones and their progenies from induced antigen signals, overcomes these hurdles and provides novel insights into the maintenance of tissue homeostasis. We demonstrate the systems use by investigating the maintenance of localised T cell tolerance in tumour immunity. In a murine tumour model, our system reveals how Tregs differentiate to a reversible, tolerance inducing state within the tumour, and recirculate, while CD8+ T cells failing to recirculate, differentiate to an increasingly exhausted, tolerant state in the tumour. These contrasting T cell behaviours provide means by which immunity can tolerate a particular anatomical niche while maintaining systemic clonal protection. Our system can thus explore lymphocyte behaviours that cannot be tracked by previous methods and will therefore provide novel insights into the fundamental mechanisms underlying immunity’s role in tissue homeostasis.
- Published
- 2023
28. Method To Visualize the Intratumor Distribution and Impact of Gemcitabine in Pancreatic Ductal Adenocarcinoma by Multimodal Imaging
- Author
-
Nicole Strittmatter, Frances M. Richards, Alan M. Race, Stephanie Ling, Daniel Sutton, Anna Nilsson, Yann Wallez, Jennifer Barnes, Gareth Maglennon, Aarthi Gopinathan, Rebecca Brais, Edmond Wong, Maria Paola Serra, James Atkinson, Aaron Smith, Joanne Wilson, Gregory Hamm, Timothy I. Johnson, Charles R. Dunlop, Brajesh P. Kaistha, Josephine Bunch, Owen J. Sansom, Zoltan Takats, Per E. Andrén, Alan Lau, Simon T. Barry, Richard J. A. Goodwin, Duncan I. Jodrell, Strittmatter, Nicole [0000-0003-1277-9608], Race, Alan M [0000-0001-8996-2641], Ling, Stephanie [0000-0002-1237-091X], Takats, Zoltan [0000-0002-0795-3467], Andrén, Per E [0000-0002-4062-7743], and Apollo - University of Cambridge Repository
- Subjects
Pancreatic Neoplasms ,Mice ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Deoxycytidine ,Multimodal Imaging ,Gemcitabine ,Carcinoma, Pancreatic Ductal ,Analytical Chemistry - Abstract
Gemcitabine (dFdC) is a common treatment for pancreatic cancer; however, it is thought that treatment may fail because tumor stroma prevents drug distribution to tumor cells. Gemcitabine is a pro-drug with active metabolites generated intracellularly; therefore, visualizing the distribution of parent drug as well as its metabolites is important. A multimodal imaging approach was developed using spatially coregistered mass spectrometry imaging (MSI), imaging mass cytometry (IMC), multiplex immunofluorescence microscopy (mIF), and hematoxylin and eosin (H&E) staining to assess the local distribution and metabolism of gemcitabine in tumors from a genetically engineered mouse model of pancreatic cancer (KPC) allowing for comparisons between effects in the tumor tissue and its microenvironment. Mass spectrometry imaging (MSI) enabled the visualization of the distribution of gemcitabine (100 mg/kg), its phosphorylated metabolites dFdCMP, dFdCDP and dFdCTP, and the inactive metabolite dFdU. Distribution was compared to small-molecule ATR inhibitor AZD6738 (25 mg/kg), which was codosed. Gemcitabine metabolites showed heterogeneous distribution within the tumor, which was different from the parent compound. The highest abundance of dFdCMP, dFdCDP, and dFdCTP correlated with distribution of endogenous AMP, ADP, and ATP in viable tumor cell regions, showing that gemcitabine active metabolites are reaching the tumor cell compartment, while AZD6738 was located to nonviable tumor regions. The method revealed that the generation of active, phosphorylated dFdC metabolites as well as treatment-induced DNA damage primarily correlated with sites of high proliferation in KPC PDAC tumor tissue, rather than sites of high parent drug abundance.
- Published
- 2022
29. Abstract PS11-18: Ceralasertib (cer) in combination with olaparib (ola) in patients (pts) with advanced breast cancer (BC): Results of phase I expansion cohorts
- Author
-
Keun-Wook Lee, Natasha Lukashchuk, Andrew Goldwin, Bienvenu Loembe, Graeme Parr, Irene Kang, Tobias Arkenau, R Roylance, Mario Campone, Claire Smith, Wassim Abida, Andrew J. Pierce, Duncan I. Jodrell, Alan Lau, Elhan Sanay, Sophie Postel-Vinay, Juanita Lopez, Sunyoung Rha, Emma Dean, Matthew G Krebs, and Seock-Ah Im
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,BRCA mutation ,Phases of clinical research ,Cancer ,Neutropenia ,medicine.disease ,Olaparib ,chemistry.chemical_compound ,Breast cancer ,Tolerability ,chemistry ,Internal medicine ,medicine ,education ,business - Abstract
Background: Alterations in BRCA1/2 are associated with ~9% of all BCs. Ola (a poly ADP-ribose polymerase inhibitor [PARPi]) is approved for treating pts with HER2-negative metastatic BC with germline BRCA mutation (gBRCAm), demonstrating an improvement in progression-free survival (PFS). Ceralasertib (an ataxia telangiectasia and Rad3-related protein ATR inhibitor) targets DNA damage repair and cell cycle regulation. Preclinical studies show synergistic antitumor effects of ola+cer vs ola monotherapy supporting the clinical evaluation of this combination. Methods: Study 4 is a modular multicenter Phase I study of cer combinations (NCT02264678). Module 2 established the Recommended Phase 2 Dose of ola+cer as ola 300 mg bid daily + cer 160 mg qd D1-7 q 28d. We report the results of two expansion cohorts testing ola+cer in pts with triple negative breast cancer with no alterations in homologous recombination repair (HRR)-related genes (HRR wt group), and pts with advanced HER2- BRCAm BC (BRCAm group), with a data cut-off of 19th June 2019. Eligible pts had to have received 1-2 prior lines of chemotherapy for metastatic disease, and were PARPi naive. Patients may have enrolled directly based on a local BRCAm test result, but all pts submitted a tumor specimen for central confirmation of a deleterious (or suspected deleterious) germline or somatic BRCA1/2 mutation. The primary objective was to investigate the safety and tolerability of the combination; secondary objectives included assessment of objective response rate by RECIST 1.1. Results: Twenty-five pts were enrolled in the HRR wt group: median age 53 (31-75), ECOG PS 0/1 13 (52%)/12 (48%), median number of prior chemotherapy lines 2 of which 8 pts (32%) had received prior platinum. Thirty-seven pts enrolled in the BRCAm group: median age 51 (24-69), ECOG PS 0/1 20 (54%)/17 (46%), median number of prior chemotherapy lines 1 of which 19 pts (51%) had received prior platinum. The most frequent all causality Adverse Events (AEs) were nausea 43 (69%), anemia 36 (58%), diarrhea 20 (32%) and vomiting 20 (32%); CTCAE ≥ Grade 3 AEs included anemia 15 (24%), neutropenia 6 (10%) with few patients discontinuing treatment. In the HRR wt group, no responses were observed, 12 (48%) pts achieved a best response of stable disease and 13 (52%) had disease progression. The median PFS was 3.1 months (80%CI 2.0,3.9). In the BRCAm group, 13 (35%) pts achieved a confirmed response (including 1 complete response), 17 (46%) stable disease and 7 (19%) pts disease progression. The median PFS in pts with a centrally confirmed BRCAm was 11.5 months (80%CI 5.8-14.8, n=30), 7.7 months (80%CI 5.8-11.4, n=37) in all pts enrolled in the BRCAm group. Eleven of the 13 responders in the BRCAm group were on study treatment for a longer duration than their treatment immediately prior to enrollment. As of 18th June 20, preliminary data shows 10 pts are still ongoing with a treatment duration ranging 20 to 35 months. Conclusion: In pts with HER2- BRCAm breast cancer, clinical efficacy was observed with durable radiological response despite adverse prognostic features in this Phase I population. The observations in this study are currently being tested in a global multicenter open-label randomised Phase 2 study: VIOLETTE (NCT03330847). Citation Format: Emma Dean, Matthew G Krebs, Seock-Ah Im, Mario Campone, Sophie Postel-Vinay, Tobias Arkenau, Juanita Lopez, Wassim Abida, Duncan Jodrell, Keun-Wook Lee, SunYoung Rha, Irene Kang, Rebecca Roylance, Graeme Parr, Claire Smith, Andrew Goldwin, Elhan Sanay, Natasha Lukashchuk, Alan Lau, Andrew Pierce, Bienvenu Loembe. Ceralasertib (cer) in combination with olaparib (ola) in patients (pts) with advanced breast cancer (BC): Results of phase I expansion cohorts [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS11-18.
- Published
- 2021
30. Defining the spatial distribution of extracellular adenosine revealed a myeloid-dependent immunosuppressive microenvironment in pancreatic ductal adenocarcinoma
- Author
-
Vincenzo Graziano, Andreas Dannhorn, Heather Hulme, Kate Williamson, Hannah Buckley, Saadia A Karim, Sheng Y. Lee, Brajesh P. Kaistha, Sabita Islam, James E. D. Thaventhiran, Frances M. Richards, Richard Goodwin, Rebecca Brais, Jennifer P Morton, Simon J. Dovedi, Alwin G. Schuller, Jim Eyles, and Duncan I. Jodrell
- Abstract
The prognosis for patients with pancreatic ductal adenocarcinoma (PDAC) remains extremely poor. It has been suggested that the adenosine pathway contributes to the ability of PDAC to evade the immune system and its resistance to immunotherapies (Immuno-Oncology Therapy, IOT), by generating extracellular adenosine (eAdo).Using genetically engineered allograft models of PDAC in syngeneic mice with differential immune infiltration and response to IOT, we showed enrichment of the adenosine pathway in tumour-infiltrating immune cells (in particular, myeloid populations). Using MS-Imaging, we showed that extracellular adenosine distribution is heterogeneous in tumours, with high concentrations in hypoxic margins that surround necrotic areas, associated with a rich myeloid infiltration, demonstrated using Imaging Mass Cytometry (IMC). Pro-tumorigenic M2 macrophages express high levels of the Adora2a receptor; particularly in the IOT resistant model. Blocking thein vivoformation and function of eAdo (Adoi), using a combination of anti-CD73 antibody and an Adora2a inhibitor slowed tumour growth and reduced metastatic burden. In addition, blocking the adenosine pathway improved the efficacy of combinations of cytotoxic agents or immunotherapy. Finally, Adoi remodelled the tumour microenvironment (TME), as evidenced by reduced infiltration of M2 macrophages and Tregs. RNAseq analysis showed that genes related to immune modulation, hypoxia and tumour stroma were downregulated following Adoi and a specific adenosine signature derived from this is associated with a poorer prognosis in patients with PDAC.The formation of eAdo appears to promote the development of the immunosuppressive TME in PDAC, contributing to its resistance to conventional and novel therapies. Therefore, inhibition of the adenosine pathway may represent a strategy to modulate the stroma and improve therapy response in patients with PDAC.
- Published
- 2022
31. Early neutrophilia marked by aerobic glycolysis sustains host metabolism and delays cancer cachexia
- Author
-
Timotheus Y.F. Halim, Shwethaa Raghunathan, Tobias Janowitz, Erwin F. Wagner, Martijn J. Schuijs, Duncan I. Jodrell, Michele Petruzzelli, Michele Vacca, Christian Frezza, Zoe Hall, David Perera, Edoardo Gaude, Miriam Ferrer, Ashok R. Venkitaraman, and Michael J. Lukey
- Subjects
business.industry ,Cancer ,medicine.disease ,Neutrophilia ,Cachexia ,Extramedullary hematopoiesis ,Anaerobic glycolysis ,Pancreatic cancer ,Cancer cell ,Cancer research ,Medicine ,Glucose homeostasis ,medicine.symptom ,business - Abstract
An elevated neutrophil-to-lymphocyte ratio negatively predicts the outcome of patients with cancer and is associated with cachexia, the terminal wasting syndrome. Here, we show using murine model systems of colorectal and pancreatic cancer that neutrophilia in the circulation and multiple organs, accompanied by extramedullary hematopoiesis, is an early event during cancer progression. Transcriptomic and metabolic assessment reveals that neutrophils in tumor-bearing animals utilize aerobic glycolysis, alike to cancer cells. Although pharmacological inhibition of aerobic glycolysis slows down tumor growth in C26 tumor-bearing mice, it precipitates cachexia, thereby shortening overall survival. This negative effect may be explained by our observation that acute depletion of neutrophils in pre-cachectic mice impairs systemic glucose homeostasis secondary to altered hepatic lipid processing. Thus, changes in neutrophil number, distribution and metabolism play an adaptive role in host metabolic homeostasis during cancer progression. Our findings provide insight into early events during cancer progression to cachexia, with implications for therapy.
- Published
- 2021
32. Quantifying Drug‐Induced Bone Marrow Toxicity Using a Novel Haematopoiesis Systems Pharmacology Model
- Author
-
Carmen Pin, Teresa Collins, Aaron Smith, S.Y. Amy Cheung, Lenka Oplustil O'Connor, James W.T. Yates, Duncan I. Jodrell, Jerome T. Mettetal, Chiara Fornari, Jodrell, Duncan [0000-0001-9360-1670], and Apollo - University of Cambridge Repository
- Subjects
Cellular differentiation ,Cell ,Biology ,Article ,Carboplatin ,Blood cell ,Bone Marrow ,medicine ,Animals ,Homeostasis ,Humans ,Pharmacology (medical) ,Progenitor cell ,Cell Proliferation ,Cell growth ,Systems Biology ,Research ,lcsh:RM1-950 ,Cell Differentiation ,Articles ,Models, Theoretical ,Hematopoiesis ,Rats ,Haematopoiesis ,medicine.anatomical_structure ,lcsh:Therapeutics. Pharmacology ,Modeling and Simulation ,Cancer research ,Bone marrow ,Systems pharmacology - Abstract
Haematological toxicity associated with cancer therapeutics is monitored by changes in blood cell count, and their primary effect is on proliferative progenitors in the bone marrow. Using observations in rat bone marrow and blood, we characterize a mathematical model that comprises cell proliferation and differentiation of the full haematopoietic phylogeny, with interacting feedback loops between lineages in homeostasis as well as following carboplatin exposure. We accurately predicted the temporal dynamics of several mature cell types related to carboplatin-induced bone marrow toxicity and identified novel insights into haematopoiesis. Our model confirms a significant degree of plasticity within bone marrow cells, with the number and type of both early progenitors and circulating cells affecting cell balance, via feedback mechanisms, through fate decisions of the multipotent progenitors. We also demonstrated cross-species translation of our predictions to patients, applying the same core model structure and considering differences in drug-dependent and physiology-dependent parameters.
- Published
- 2019
33. Abstract 6103: Reshaping the myeloid-dependent pro-tumorigenic microenvironment in PDAC by targeting the extracellular adenosine pathway: A therapeutic opportunity
- Author
-
Vincenzo Graziano, Andreas Dannhorn, Kate Williamson, Heather Hulme, Hannah Buckley, Sheng Y. Lee, Sabita Islam, James E. Thaventhiran, Richard Goodwin, Rebecca Brais, Simon J. Dovedi, Alwin Schuller, Jim Eyles, and Duncan I. Jodrell
- Subjects
Cancer Research ,Oncology - Abstract
The prognosis for patients with pancreatic adenocarcinoma (PDAC) remains extremely poor. PDAC is resistant to both conventional therapies and emerging immunotherapies (IOT), apart from tumors with mismatch repair deficiency. This may be explained in part by its low tumor mutational burden (TMB) but also by its immunosuppressive tumor microenvironment (TME). It has been suggested that CD73, a member of the adenosine pathway, expressed on cancer cells contributes to immune escape and resistance to cytotoxic/radiotherapy treatment. The adenosine pathway converts the immune activator ATP, released by dying cells during cell turnover or after treatment, to extracellular Adenosine (eAdo), which is immunosuppressive. Using syngeneic, in vivo models by s.c. implantation of KPC-derived cell lines (courtesy of Ben Stanger, UPenn) with differential immune infiltration and response to IOT [resistant (IOTResi) or responsive (IOTResp)], we showed by flow cytometry that the adenosine pathway is enriched in the tumor-infiltrating immune cells (in particular myeloid populations) which co-express CD39 and CD73, enabling the formation of eAdo. Mass Spec Imaging (MSI) revealed that adenosine distribution is heterogeneous in the tumors with high concentrations in the hypoxic margins that surround necrotic areas. Subpopulations of myeloid cells infiltrating the lesions are a target for eAdo, expressing high levels of adenosine receptor Adora2a. We discovered that pro-tumorigenic M2 macrophages have the highest expression of the receptor and significantly higher in the IOTResi model. Blocking the in vivo formation and function of eAdo in IOTResi tumors, using a combination of anti-CD73 antibody (2C5, murine IgG1-Fc) and an inhibitor of Adora2a (AZD4635) reduced the presence of eAdo, slowed tumor growth and reduced the lung metastatic burden. The combination remodeled the TME, reducing the infiltration by M2 macrophages, particularly those that are PD-L1 positive and diminished the frequency of infiltrating Tregs. Bulk RNAseq analysis demonstrated a profound dependency of the TME on the presence of eAdo. Genes related to cytokine/chemokine signaling, immunosuppression/inflammation, hypoxia, metastasis and collagen production are strongly downregulated following administration of anti-CD73Ab/Adora2ai. In addition, blocking the adenosine pathway improved the efficacy of combinations of cytotoxics (gemcitabine/ATR inhibitor) and immunotherapy (aCD40/anti-PDL1Ab). The formation of eAdo appears to be a factor in the development of the immunosuppressive TME in PDAC, contributing to its resistance to conventional and novel therapies. Therefore, inhibition of the adenosine pathway using a CD73Ab and an Adora2ai may represent a strategy to modulate the PDAC stroma and improve therapy response in patients with PDAC. Citation Format: Vincenzo Graziano, Andreas Dannhorn, Kate Williamson, Heather Hulme, Hannah Buckley, Sheng Y. Lee, Sabita Islam, James E. Thaventhiran, Richard Goodwin, Rebecca Brais, Simon J. Dovedi, Alwin Schuller, Jim Eyles, Duncan I. Jodrell. Reshaping the myeloid-dependent pro-tumorigenic microenvironment in PDAC by targeting the extracellular adenosine pathway: A therapeutic opportunity [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 6103.
- Published
- 2022
34. Scheduling nab-paclitaxel combined with gemcitabine as first-line treatment for metastatic pancreatic adenocarcinoma
- Author
-
Richard J. Ellis, Harpreet Wasan, Karen McAdam, S. Arif, Lisa Bax, Roopinder Gillmore, Jonathan Wadsley, Duncan I. Jodrell, Sebastian Cummins, Albrecht Neesse, Pippa Corrie, Yuk Ting Ma, Daniel H. Palmer, Rebecca Brais, J. Evans, David Propper, Aarthi Gopinathan, A. Chhabra, Martin Scott-Brown, R. Skells, Andrea Machin, K. Dalchau, A. Dayim, P. Bundi, Christopher Isherwood, Bristi Basu, C. Lwuji, John Bridgewater, David A. Tuveson, Alan Anthoney, Lucy Wall, S Falk, Juan W. Valle, Wendi Qian, Valle, J. W. [0000-0002-1999-0863], Bridgewater, J. [0000-0001-9186-1604], Apollo - University of Cambridge Repository, Valle, JW [0000-0002-1999-0863], and Bridgewater, J [0000-0001-9186-1604]
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Paclitaxel ,Deoxycytidine ,Drug Administration Schedule ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Albumins ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Carcinoma ,Chemotherapy ,Humans ,Progression-free survival ,631/67/1504/1713 ,631/67/1059/99 ,030304 developmental biology ,Aged ,Aged, 80 and over ,0303 health sciences ,Manchester Cancer Research Centre ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Hazard ratio ,article ,Pancreatic cancer ,Metastatic Pancreatic Adenocarcinoma ,Middle Aged ,medicine.disease ,Gemcitabine ,Confidence interval ,Progression-Free Survival ,Clinical trial ,Pancreatic Neoplasms ,030220 oncology & carcinogenesis ,Concomitant ,Female ,business ,medicine.drug ,Carcinoma, Pancreatic Ductal - Abstract
Background Nab-paclitaxel plus gemcitabine (nabP+gemcitabine) offers modest survival gains for patients with metastatic pancreatic ductal adenocarcinoma (PDAC). Sequential scheduling of nabP+gemcitabine in a PDAC mouse model improved efficacy; this hypothesis was tested in a clinical trial. Methods Patients with previously untreated metastatic PDAC were randomised to receive nabP+gemcitabine administered either concomitantly on the same day, or sequentially, with gemcitabine administered 24 h after nabP. The primary outcome measure was progression-free survival (PFS). Secondary outcome measures were objective response rate (ORR), overall survival (OS), safety, quality of life (QoL) and predictive biomarkers. Results In total, 71 patients received sequential (SEQ) and 75 concomitant (CON) treatment. Six-month PFS was 46% with SEQ and 32% with CON scheduling. Median PFS (5.6 versus 4.0 months, hazard ratio [HR] 0.67, 95% confidence interval [95% CI] 0.47–0.95, p = 0.022) and ORR (52% versus 31%, p = 0.023) favoured the SEQ arm; median OS was 10.2 versus 8.2 months (HR 0.93, 95% CI 0.65–1.33, p = 0.70). CTCAE Grade ≥3 neutropaenia incidence doubled with SEQ therapy but was not detrimental to QoL. Strongly positive tumour epithelial cytidine deaminase (CDA) expression favoured benefit from SEQ therapy (PFS HR 0.31, 95% CI 0.13–0.70). Conclusions SEQ delivery of nabP+gemcitabine improved PFS and ORR, with manageable toxicity, but did not significantly improve OS. Clinical trial registration ISRCTN71070888; ClinialTrials.gov (NCT03529175).
- Published
- 2021
- Full Text
- View/download PDF
35. Mechanisms Underlying Vascular Endothelial Growth Factor Receptor Inhibition-Induced Hypertension – The Hypaz Trial
- Author
-
Ian B. Wilkinson, Carola-Bibiane Schönlieb, Sara Santos Franco, Yoeri E. Boink, Christoph Brune, Annette Hubsch, Adam McGeoch, Lucy Yang, Fraz Mir, Simon Bond, Carmel M. McEniery, Nicola Ramenatte, Duncan I. Jodrell, Kaisa M. Mäki-Petäjä, Gayathri Anandappa, Joseph Cheriyan, Paul A R Meyer, Parag R Gajendragadkar, Biomedical Photonic Imaging, Applied Analysis, McEniery, Carmel [0000-0003-3636-0705], Bond, Simon [0000-0003-2528-1040], Wilkinson, Ian [0000-0001-6598-9399], Jodrell, Duncan [0000-0001-9360-1670], Cheriyan, Joseph [0000-0001-6921-1592], and Apollo - University of Cambridge Repository
- Subjects
Male ,Angiogenesis Inhibitors ,030204 cardiovascular system & hematology ,chemistry.chemical_compound ,0302 clinical medicine ,cardiovascular disease ,Neoplasms ,pazopanib ,Cardiac Output ,ComputingMilieux_MISCELLANEOUS ,Aged, 80 and over ,Sulfonamides ,biology ,Microvascular Density ,Middle Aged ,Vascular endothelial growth factor ,Nitric oxide synthase ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Hypertension ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Indazoles ,Diastole ,Urology ,Article ,Pazopanib ,03 medical and health sciences ,Vascular Stiffness ,Internal Medicine ,medicine ,Humans ,Aged ,business.industry ,microvascular rarefaction ,Hemodynamics ,acetylcholine ,Blood pressure ,Pyrimidines ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Vascular resistance ,biology.protein ,Microvascular Rarefaction ,Vascular Resistance ,Endothelium, Vascular ,business - Abstract
Drugs targeting the VEGF (vascular endothelial growth factor) signaling pathway are approved for several malignancies. Unfortunately, VEGF inhibitors lead to hypertension in 30% to 80% patients. Reduced nitric oxide synthase activity, microvascular rarefaction, and increased vascular resistance have been proposed as potential mechanisms. We aimed to assess these mechanisms in patients receiving the VEGF inhibitor, pazopanib, for cancer. Twenty-seven normotensive patients with advanced solid malignancies received pazopanib 800 mg od. Endothelial function was assessed using forearm plethysmography with intraarterial infusions of acetylcholine. Detailed hemodynamic measurements were taken. Density and diameter of the conjunctival and episcleral microvasculature were evaluated using hemoglobin video imaging. Measurements were taken at baseline, 2, and 12 weeks after initiation of pazopanib or earlier if patients became hypertensive. By the end of the trial, systolic blood pressure increased by 12 mm Hg (95% CI, 4–19 mm Hg; P =0.003), diastolic by 10 mm Hg (95% CI, 5–15 mm Hg; P 5 (95% CI, 616–1168 dynes×s/cm 5 ; P P P =0.003) and diameter by −2.09 µm (95% CI, −3.95 to −0.19 µm; P =0.03). A post hoc colorimetric assay revealed that pazopanib inhibited acetylcholinesterase activity by −56% (95% CI, −62% to −52%; P Registration: URL: https://www.clinicaltrials.gov ; Unique identifier: NCT01392352.
- Published
- 2021
36. Quantifying cell cycle-dependent drug sensitivities in cancer using a high throughput synchronisation and screening approach
- Author
-
Alan Lau, Frances M. Richards, Christopher J. Minteer, Sandra Bernaldo de Quirós Fernández, Timothy Isaac Johnson, Duncan I. Jodrell, Daniel Kottmann, Charles R. Dunlop, Yann Wallez, Larissa S. Carnevalli, Kottmann, Daniel [0000-0002-1750-4016], Carnevalli, Larissa S [0000-0001-7432-0195], Apollo - University of Cambridge Repository, and Jodrell, Duncan [0000-0001-9360-1670]
- Subjects
0301 basic medicine ,Medicine (General) ,Time Factors ,Pyridines ,Cell ,synchronisation ,Cell Culture Techniques ,Deoxycytidine ,Piperazines ,0302 clinical medicine ,Neoplasms ,media_common ,Cancer ,Nocodazole ,Cell Cycle ,Drug Synergism ,General Medicine ,Cell cycle ,Tubulin Modulators ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,combination treatment ,MCF-7 Cells ,Medicine ,medicine.drug ,Drug ,Cell Survival ,media_common.quotation_subject ,High-throughput screening ,Computational biology ,Biology ,Palbociclib ,General Biochemistry, Genetics and Molecular Biology ,Small Molecule Libraries ,03 medical and health sciences ,R5-920 ,Cell Line, Tumor ,medicine ,Humans ,Mitosis ,Cell Proliferation ,medicine.disease ,Gemcitabine ,High-Throughput Screening Assays ,030104 developmental biology ,Commentary ,Drug Screening Assays, Antitumor ,high throughput screening ,HeLa Cells - Abstract
BackgroundChemotherapy and targeted agent anti-cancer efficacy is largely dependent on the proliferative state of tumours, as exemplified by agents that target DNA synthesis/replication or mitosis. As a result, cell cycle specificities of a number of cancer drugs are well known. However, they are yet to be described in a quantifiable manner.MethodsA scalable cell synchronisation protocol used to screen a library of 235 anti-cancer compounds exposed over six hours in G1 or S/G2 accumulated AsPC-1 cells to generate a cell cycle specificity (CCS) score.FindingsThe synchronisation method was associated with reduced method-related cytotoxicity compared to nocodazole, delivering sufficient cell cycle purity and cell numbers to run high-throughput drug library screens. Compounds were identified with G1 and S/G2-associated specificities that, overall, functionally matched with a compound's target/mechanism of action. This annotation was used to describe a synergistic schedule using the CDK4/6 inhibitor, palbociclib, prior to gemcitabine/AZD6738 as well as describe the correlation between the CCS score and published synergistic/antagonistic drug schedules.InterpretationThis is the first highly quantitative description of cell cycle-dependent drug sensitivities that utilised a tractable and tolerated method with potential uses outside the present study. Drug treatments such as those shown to be G1 or S/G2 associated may benefit from scheduling considerations such as after CDK4/6 inhibitors and being first in drug sequences respectively.FundingCancer Research UK (CRUK) Institute core grants C14303/A17197 and C9545/A29580. The Li Ka Shing Centre where this work was performed was generously funded by CK Hutchison Holdings Limited, the University of Cambridge, CRUK, The Atlantic Philanthropies and others.
- Published
- 2021
37. CXCR4 inhibition in human pancreatic and colorectal cancers induces an integrated immune response
- Author
-
T. Isaac Johnson, Zhikai Wang, Claire M. Connell, James Thaventhiran, Ferdia A. Gallagher, Tobias Janowitz, Duncan I. Jodrell, Martin Smoragiewicz, Petros Barmpounakis, Lukasz Magiera, Douglas T. Fearon, Daniele Biasci, Irena Hudecova, Edmund Godfrey, Davina Gale, Nitzan Rosenfeld, Bristi Basu, Aarthi Gopinathan, Frances M. Richards, Lisa Bax, Maria Pawula, Rebecca Brais, Fraz Mir, Christopher Isherwood, Elizabeta C. Popa, Ya Gao, Smoragiewicz, Martin [0000-0002-4934-6323], Connell, Claire M [0000-0002-6696-8415], Wang, Zhikai [0000-0002-8565-6011], Thaventhiran, James ED [0000-0001-8616-074X], Gallagher, Ferdia A [0000-0003-4784-5230], Barmpounakis, Petros [0000-0002-1020-3886], Richards, Frances M [0000-0001-7947-7853], Janowitz, Tobias [0000-0002-7820-3727], and Apollo - University of Cambridge Repository
- Subjects
Male ,CCR2 ,Benzylamines ,Medical Sciences ,medicine.medical_treatment ,pancreatic cancer ,CXCR3 ,Cyclams ,CXCR4 ,CXCR5 ,Receptors, Interleukin-8A ,Chemokine receptor ,Heterocyclic Compounds ,Tumor Microenvironment ,Medicine ,Multidisciplinary ,Biological Sciences ,Middle Aged ,Hematopoietic Stem Cell Mobilization ,medicine.anatomical_structure ,Female ,Immunotherapy ,Colorectal Neoplasms ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,Receptors, CXCR5 ,Receptors, CXCR4 ,Receptors, CXCR3 ,Receptors, CCR2 ,T cell ,chemical and pharmacologic phenomena ,colorectal cancer ,AMD3100 ,Immune system ,Pancreatic cancer ,Humans ,Pancreas ,Aged ,Receptors, CXCR6 ,Tumor microenvironment ,business.industry ,Comment ,Immunity ,medicine.disease ,Chemokine CXCL12 ,Pancreatic Neoplasms ,Cancer research ,business - Abstract
Significance Patients with microsatellite stable (MSS) pancreatic (PDA) or colorectal cancer (CRC) do not respond to immunotherapy with inhibitors of T cell checkpoints. A possible explanation is suggested by finding that cancer cells in these tumors are coated with the chemokine, CXCL12, and that stimulation of CXCR4, the CXCL12 receptor on immune cells, suppresses directed migration mediated by other chemokine receptors on these cells. We assessed the relevance of these findings by treating patients for seven days with continuous infusion of AMD3100/Plerixafor, a CXCR4 inhibitor. Comparison of pre- and end-of-treatment paired biopsies of metastatic lesions by transcriptomic analysis revealed that AMD3100 induced an integrated immune response that is predictive of a clinical response to T cell checkpoint inhibition., Inhibition of the chemokine receptor CXCR4 in combination with blockade of the PD-1/PD-L1 T cell checkpoint induces T cell infiltration and anticancer responses in murine and human pancreatic cancer. Here we elucidate the mechanism by which CXCR4 inhibition affects the tumor immune microenvironment. In human immune cell-based chemotaxis assays, we find that CXCL12-stimulated CXCR4 inhibits the directed migration mediated by CXCR1, CXCR3, CXCR5, CXCR6, and CCR2, respectively, chemokine receptors expressed by all of the immune cell types that participate in an integrated immune response. Inhibiting CXCR4 in an experimental cancer medicine study by 1-wk continuous infusion of the small-molecule inhibitor AMD3100 (plerixafor) induces an integrated immune response that is detected by transcriptional analysis of paired biopsies of metastases from patients with microsatellite stable colorectal and pancreatic cancer. This integrated immune response occurs in three other examples of immune-mediated damage to noninfected tissues: Rejecting renal allografts, melanomas clinically responding to anti-PD1 antibody therapy, and microsatellite instable colorectal cancers. Thus, signaling by CXCR4 causes immune suppression in human pancreatic ductal adenocarcinoma and colorectal cancer by impairing the function of the chemokine receptors that mediate the intratumoral accumulation of immune cells.
- Published
- 2020
38. Complete loss of ATM function augments replication catastrophe induced by ATR inhibition and gemcitabine in pancreatic cancer models
- Author
-
Sandra Bernaldo de Quirós Fernández, Elaine Cadogan, Charles R. Dunlop, Alan Lau, Frances M. Richards, Timothy Isaac Johnson, Stephen T. Durant, Duncan I. Jodrell, Yann Wallez, Dunlop, Charles R. [0000-0002-8194-2598], Jodrell, Duncan I. [0000-0001-9360-1670], Apollo - University of Cambridge Repository, Dunlop, Charles R [0000-0002-8194-2598], and Jodrell, Duncan I [0000-0001-9360-1670]
- Subjects
Cancer Research ,Indoles ,endocrine system diseases ,Pyridines ,Ataxia Telangiectasia Mutated Proteins ,631/154/53/2423 ,Deoxycytidine ,631/80/86/2371 ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,RNA, Small Interfering ,631/67/1504/1713 ,631/67/1059/99 ,0303 health sciences ,Sulfonamides ,medicine.diagnostic_test ,article ,Drug Synergism ,631/67/1059/602 ,Oncology ,Checkpoint signalling ,030220 oncology & carcinogenesis ,Sulfoxides ,Quinolines ,Female ,medicine.drug ,Carcinoma, Pancreatic Ductal ,Programmed cell death ,Pancreatic ductal adenocarcinoma ,Mice, 129 Strain ,Morpholines ,Immunofluorescence ,Predictive markers ,Flow cytometry ,03 medical and health sciences ,Targeted therapies ,In vivo ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Chemotherapy ,Animals ,Humans ,030304 developmental biology ,Cell Proliferation ,business.industry ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,Pyrimidines ,Cancer research ,business ,Function (biology) - Abstract
BackgroundPersonalised medicine strategies may improve outcomes in pancreatic ductal adenocarcinoma (PDAC), but validation of predictive biomarkers is required. Having developed a clinical trial to assess the ATR inhibitor, AZD6738, in combination with gemcitabine (ATRi/gem), we investigated ATM loss as a predictive biomarker of response to ATRi/gem in PDAC.MethodsThrough kinase inhibition, siRNA depletion and CRISPR knockout of ATM, we assessed how ATM targeting affected the sensitivity of PDAC cells to ATRi/gem. Using flow cytometry, immunofluorescence and immunoblotting, we investigated how ATRi/gem synergise in ATM-proficient and ATM-deficient cells, before assessing the impact of ATM loss on ATRi/gem sensitivity in vivo.ResultsComplete loss of ATM function (through pharmacological inhibition or CRISPR knockout), but not siRNA depletion, sensitised to ATRi/gem. In ATM-deficient cells, ATRi/gem-induced replication catastrophe was augmented, while phospho-Chk2-T68 and phospho-KAP1-S824 persisted via DNA-PK activity. ATRi/gem caused growth delay in ATM-WT xenografts in NSG mice and induced regression in ATM-KO xenografts.ConclusionsATM loss augments replication catastrophe-mediated cell death induced by ATRi/gem and may predict clinical responsiveness to this combination. ATM status should be carefully assessed in tumours from patients with PDAC, since distinction between ATM-low and ATM-null could be critical in maximising the success of clinical trials using ATM expression as a predictive biomarker.
- Published
- 2020
- Full Text
- View/download PDF
39. Targeting of 5-aza-2'-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme
- Author
-
Kenneth G. MacLeod, Bernard H Ramsahoye, Katan Patel, Duncan I. Jodrell, Jacqueline Dickson, Shahida Din, Jodrell, Duncan [0000-0001-9360-1670], and Apollo - University of Cambridge Repository
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Proteasome Endopeptidase Complex ,Biology ,Gene Regulation, Chromatin and Epigenetics ,Decitabine ,environment and public health ,Cell Line ,DNA Methyltransferase 3A ,chemistry.chemical_compound ,Mice ,Aphidicolin ,MG132 ,Genetics ,medicine ,Animals ,Ligase activity ,DNA (Cytosine-5-)-Methyltransferases ,Enzyme Inhibitors ,DNA synthesis ,urogenital system ,DNA ,DNA Methylation ,Molecular biology ,Chromatin ,CpG site ,chemistry ,DNA methylation ,embryonic structures ,Proteasome inhibitor ,Azacitidine ,medicine.drug - Abstract
5-Aza-2'-deoxycytidine (5-aza-dC) is a nucleoside analogue with cytotoxic and DNA demethylating effects. Here we show that 5-aza-dC induces the proteasomal degradation of free (non-chromatin bound) DNMT1 through a mechanism which is dependent on DNA synthesis and the targeting of incorporated 5-aza-dC residues by DNMT1 itself. Thus, 5-aza-dC induces Dnmt1 degradation in wild-type mouse ES cells, but not in Dnmt [3a(-/-), 3b(-/-)] mouse ES cells which express Dnmt1 but lack DNA methylation (< 0.7% of CpG methylated) and contain few hemi-methylated CpG sites, these being the preferred substrates for Dnmt1. We suggest that adducts formed between DNMT1 and 5-aza-dC molecules in DNA induce a ubiquitin-E3 ligase activity which preferentially targets free DNMT1 molecules for degradation by the proteasome. The proteasome inhibitor MG132 prevents DNMT1 degradation and reduces hypomethylation induced by 5-aza-dC.
- Published
- 2020
- Full Text
- View/download PDF
40. In vitro and in vivo activity and cross resistance profiles of novel ruthenium (II) organometallic arene complexes in human ovarian cancer
- Author
-
Haimei Chen, Peter J. Sadler, Duncan I. Jodrell, A.A. Ritchie, M. Muir, Jeffrey Cummings, Morris Robert Edward, R E Aird, Jodrell, Duncan [0000-0001-9360-1670], and Apollo - University of Cambridge Repository
- Subjects
Cancer Research ,Ligands ,Carboplatin ,chemistry.chemical_compound ,Mice ,NAMI-A ,Ovarian Neoplasms ,structure activity relationships ,Biological activity ,Calcium Channel Blockers ,Drug Resistance, Multiple ,Ruthenium ,Neoplasm Proteins ,ruthenium(II) ,Oncology ,Azacitidine ,Ruthenium Compounds ,Female ,medicine.drug ,Antimetabolites, Antineoplastic ,Stereochemistry ,chemistry.chemical_element ,Mice, Nude ,Antineoplastic Agents ,Biology ,Decitabine ,Structure-Activity Relationship ,In vivo ,medicine ,Organometallic Compounds ,Structure–activity relationship ,Animals ,Humans ,Experimental Therapeutics ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Edetic Acid ,Cisplatin ,drug resistance ,Ligand ,organometallic arene complexes ,ovarian xenografts ,Xenograft Model Antitumor Assays ,chemistry ,Verapamil ,Drug Resistance, Neoplasm ,Drug Design ,Immunology ,Drug Screening Assays, Antitumor - Abstract
Ruthenium complexes offer the potential of reduced toxicity, a novel mechanism of action, non-cross resistance and a different spectrum of activity compared to platinum containing compounds. Thirteen novel ruthenium(II) organometallic arene complexes have been evaluated for activity (in vitro and in vivo) in models of human ovarian cancer, and cross-resistance profiles established in cisplatin and multi-drug-resistant variants. A broad range of IC50 values was obtained (0.5 to >100 μM) in A2780 parental cells with two compounds (RM175 and HC29) equipotent to carboplatin (6 μM), and the most active compound (HC11) equipotent to cisplatin (0.6 μM). Stable bi-dentate chelating ligands (ethylenediamine), a more hydrophobic arene ligand (tetrahydroanthracene) and a single ligand exchange centre (chloride) were associated with increased activity. None of the six active ruthenium(II) compounds were cross-resistant in the A2780cis cell line, demonstrated to be 10-fold resistant to cisplatin/carboplatin by a mechanism involving, at least in part, silencing of MLH1 protein expression via methylation. Varying degrees of cross-resistance were observed in the P-170 glycoprotein overexpressing multi-drug-resistant cell line 2780AD that could be reversed by co-treatment with verapamil. In vivo activity was established with RM175 in the A2780 xenograft together with non-cross-resistance in the A2780cis xenograft and a lack of activity in the 2780AD xenograft. High activity coupled to non cross-resistance in cisplatin resistant models merit further development of this novel group of anticancer compounds. British Journal of Cancer (2002) 86, 1652–1657. DOI: 10.1038/sj/bjc/6600290 www.bjcancer.com © 2002 Cancer Research UK
- Published
- 2020
- Full Text
- View/download PDF
41. Opportunities for Quantitative Translational Modeling in Oncology
- Author
-
Rajesh Jena, Emma Martin, Carina Dunlop, James W.T. Yates, Duncan I. Jodrell, Sonya C. Chapman, Paolo Vicini, Juan A. Delgado-SanMartin, Helen Byrne Byrne, Giovanni Y. Di Veroli, Lourdes Cucurull-Sanchez, Herbert Struemper, Antonio Ramos-Montoya, Francois Mercier, Simon J. Dovedi, and Tao Chen
- Subjects
Pharmacology ,Oncology ,medicine.medical_specialty ,Concentration dependence ,Psychological intervention ,MEDLINE ,030226 pharmacology & pharmacy ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Drug development ,Quantitative pharmacology ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Pharmacology (medical) ,Psychology ,Systems pharmacology - Abstract
A 2‐day meeting was held by members of the UK Quantitative Systems Pharmacology Network (http://www.qsp‐uk.net/) in November 2018 on the topic of Translational Challenges in Oncology. Participants from a wide range of backgrounds were invited to discuss current and emerging modeling applications in nonclinical and clinical drug development, and to identify areas for improvement. This resulting perspective explores opportunities for impactful quantitative pharmacology approaches. Four key themes arose from the presentations and discussions that were held, leading to the following recommendations: • Evaluate the predictivity and reproducibility of animal cancer models through precompetitive collaboration. • Apply mechanism of action (MoA) based mechanistic models derived from nonclinical data to clinical trial data. • Apply MoA reflective models across trial data sets to more robustly quantify the natural history of disease and response to differing interventions. • Quantify more robustly the dose and concentration dependence of adverse events through mathematical modelling techniques and modified trial design.
- Published
- 2020
42. Depletion of Macrophages Improves Therapeutic Response to Gemcitabine in Murine Pancreas Cancer
- Author
-
Soeren M, Buchholz, Robert G, Goetze, Shiv K, Singh, Christoph, Ammer-Herrmenau, Frances M, Richards, Duncan I, Jodrell, Malte, Buchholz, Patrick, Michl, Volker, Ellenrieder, Elisabeth, Hessmann, and Albrecht, Neesse
- Subjects
Communication ,pancreatic cancer ,drug delivery ,chemoresistance ,macrophages - Abstract
Background: The tumor microenvironment (TME) is composed of fibro-inflammatory cells and extracellular matrix (ECM) components. However, the exact contribution of the various TME compartments towards therapeutic response is unknown. Here, we aim to dissect the specific contribution of tumor-associated macrophages (TAMs) towards drug delivery and response in pancreatic ductal adenocarcinoma (PDAC). Methods: The effect of gemcitabine was assessed in human and murine macrophages, human pancreatic stellate cells (hPSCs), and tumor cells (L3.6pl, BxPC3 and KPC) in vitro. The drug metabolism of gemcitabine was analyzed by liquid chromatography–tandem mass spectrometry (LC–MS/MS). Preclinical studies were conducted using KrasG12D;p48-Cre and KrasG12D;p53172H;Pdx-Cre mice to investigate gemcitabine delivery at different stages of tumor progression and upon pharmacological TAM depletion. Results: Gemcitabine accumulation was significantly increased in murine PDAC tissue compared to pancreatic intraepithelial neoplasia (PanIN) lesions and healthy control pancreas tissue. In vitro, macrophages accumulated and rapidly metabolized gemcitabine resulting in a significant drug scavenging effect for gemcitabine. Finally, pharmacological TAM depletion enhanced therapeutic response to gemcitabine in tumor-bearing KPC mice. Conclusion: Macrophages rapidly metabolize gemcitabine in vitro, and pharmacological depletion improves the therapeutic response to gemcitabine in vivo. Our study supports the notion that TAMs might be a promising therapeutic target in PDAC.
- Published
- 2020
43. Paclitaxel and CYC3, an aurora kinase A inhibitor, synergise in pancreatic cancer cells but not bone marrow precursor cells
- Author
-
Yao Lin, A. Robertson, Duncan I. Jodrell, Jo L. Bramhall, Jennifer Harrington, Daniella Zheleva, Tashinga E. Bapiro, Frances M. Richards, B-F Krippendorff, Richards, Fran [0000-0001-7947-7853], Jodrell, Duncan [0000-0001-9360-1670], and Apollo - University of Cambridge Repository
- Subjects
Cancer Research ,Paclitaxel ,pancreatic cancer ,Bone Marrow Cells ,Pharmacology ,Protein Serine-Threonine Kinases ,Granulocyte-Macrophage Progenitor Cells ,chemistry.chemical_compound ,Aurora Kinases ,Precursor cell ,Pancreatic cancer ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Protein Kinase Inhibitors ,Aurora Kinase A ,Protein-Serine-Threonine Kinases ,business.industry ,Stem Cells ,Drug Synergism ,medicine.disease ,Pancreatic Neoplasms ,medicine.anatomical_structure ,Oncology ,chemistry ,embryonic structures ,Cancer research ,combination treatment ,Bone marrow ,biological phenomena, cell phenomena, and immunity ,Stem cell ,business ,Translational Therapeutics ,CYC3 ,mitotic inhibitor - Abstract
BACKGROUND: Amplification of aurora kinase A (AK-A) overrides the mitotic spindle assembly checkpoint, inducing resistance to taxanes. RNA interference targeting AK-A in human pancreatic cancer cell lines enhanced taxane chemosensitivity. In this study, a novel AK-A inhibitor, CYC3, was investigated in pancreatic cancer cell lines, in combination with paclitaxel. METHODS: Western blot, flow cytometry and immunostaining were used to investigate the specificity of CYC3. Sulforhodamine B staining, time-lapse microscopy and colony-formation assays were employed to evaluate the cytotoxic effect of CYC3 and paclitaxel. Human colony-forming unit of granulocyte and macrophage (CFU-GM) cells were used to compare the effect in tumour and normal tissue. RESULTS: CYC3 was shown to be a specific AK-A inhibitor. Three nanomolar paclitaxel (growth inhibition 50% (GI(50)) 3 nM in PANC-1, 5.1 nM in MIA PaCa-2) in combination with 1 μM CYC3 (GI(50) 1.1 μM in MIA PaCa2 and 2 μM in PANC-1) was synergistic in inhibiting pancreatic cell growth and causing mitotic arrest, achieving similar effects to 10-fold higher concentrations of paclitaxel (30 nM). In CFU-GM cells, the effect of the combination was simply additive, displaying significantly less myelotoxicity compared with high concentrations of paclitaxel (30 nM; 60-70% vs 100% inhibition). CONCLUSION: The combination of lower doses of paclitaxel and CYC3 merits further investigation with the potential for an improved therapeutic index in vivo.
- Published
- 2020
- Full Text
- View/download PDF
44. Early relapse on adjuvant gemcitabine associated with an exceptional response to 2nd line capecitabine chemotherapy in a patient with pancreatic adenosquamous carcinoma with strong intra-tumoural expression of cytidine deaminase: a case report
- Author
-
Jane Risdall, Tobias Janowitz, Duncan I. Jodrell, Sara Upponi, Claire M. Connell, Hayley C. Whitaker, Rebecca Brais, Ian Beh, Pippa Corrie, Jodrell, Duncan I. [0000-0001-9360-1670], Apollo - University of Cambridge Repository, and Jodrell, Duncan I [0000-0001-9360-1670]
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Medical and radiation oncology ,Adenosquamous carcinoma ,medicine.medical_treatment ,Gene Expression ,Pancreatic Adenosquamous Carcinoma ,Exceptional Response ,Case Report ,lcsh:RC254-282 ,Deoxycytidine ,Capecitabine ,Carcinoma, Adenosquamous ,Surgical oncology ,Recurrence ,Internal medicine ,Genetics ,Medicine ,Humans ,ComputingMilieux_MISCELLANEOUS ,Chemotherapy ,Drug metabolism ,Cytidine deaminase ,business.industry ,Pancreatic adenosquamous carcinoma ,Middle Aged ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Magnetic Resonance Imaging ,Gemcitabine ,Pancreatic Neoplasms ,Treatment Outcome ,Chemotherapy, Adjuvant ,ComputerApplications_GENERAL ,Retreatment ,Female ,business ,Tomography, X-Ray Computed ,medicine.drug - Abstract
Funder: Cambridge Experimental Medicine Initiative, Funder: Cancer Research United Kingdom, Background: Pancreatic adenosquamous carcinoma has a poor prognosis, with limited prospective trial data to guide optimal treatment. The potential impact of drug metabolism on the treatment response of patients with pancreatic adenosquamous carcinoma is largely unknown. Case presentation: We describe the case of a 51 year old woman with pancreatic adenosquamous carcinoma who, following surgical resection, experienced early disease relapse during adjuvant gemcitabine therapy. Paradoxically, this was followed by an exceptional response to capecitabine therapy lasting 34.6 months. Strong expression of cytidine deaminase was detected within the tumour. Conclusions: This case study demonstrates that early relapse during adjuvant chemotherapy for pancreatic adenosquamous carcinoma may be compatible with a subsequent exceptional response to second line chemotherapy, an important observation given the poor overall prognosis of patients with adenosquamous carcinoma. Cytidine deaminase is predicted to inactivate gemcitabine and, conversely, catalyze capecitabine activation. We discuss strong intra-tumoural expression of cytidine deaminase as a potential mechanism to explain this patient’s disparate responses to gemcitabine and capecitabine therapy, and highlight the benefit that may be gained from considering similar determinants of response to chemotherapy in clinical practice.
- Published
- 2020
45. The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression
- Author
-
James W.T. Yates, Yann Wallez, Charles R. Dunlop, Frances M. Richards, Chiara Fornari, Siang-Boon Koh, Timothy Isaac Johnson, Duncan I. Jodrell, Alan Lau, and Sandra Bernaldo de Quirós Fernández
- Subjects
0301 basic medicine ,Cancer Research ,Cell cycle checkpoint ,medicine.drug_class ,Kinase ,business.industry ,medicine.disease ,Antimetabolite ,Gemcitabine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,In vivo ,Ataxia-telangiectasia ,medicine ,Cancer research ,Growth inhibition ,Clonogenic assay ,business ,medicine.drug - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers, and overall survival rates have barely improved over the past five decades. The antimetabolite gemcitabine remains part of the standard of care but shows very limited antitumor efficacy. Ataxia telangiectasia and Rad3-related protein (ATR), the apical kinase of the intra–S-phase DNA damage response, plays a central role in safeguarding cells from replication stress and can therefore limit the efficacy of antimetabolite drug therapies. We investigated the ability of the ATR inhibitor, AZD6738, to prevent the gemcitabine-induced intra–S-phase checkpoint activation and evaluated the antitumor potential of this combination in vitro and in vivo. In PDAC cell lines, AZD6738 inhibited gemcitabine-induced Chk1 activation, prevented cell-cycle arrest, and restrained RRM2 accumulation, leading to the strong induction of replication stress markers only with the combination. Moreover, synergistic growth inhibition was identified in a panel of 5 mouse and 7 human PDAC cell lines using both Bliss Independence and Loewe models. In clonogenic assays, the combination abrogated survival at concentrations for which single agents had minor effects. In vivo, AZD6738 in combination with gemcitabine was well tolerated and induced tumor regression in a subcutaneous allograft model of a KrasG12D; Trp53R172H; Pdx-Cre (KPC) mouse cancer cell line, significantly extending survival. Remarkably, the combination also induced regression of a subgroup of KPC autochthonous tumors, which generally do not respond well to conventional chemotherapy. Altogether, our data suggest that AZD6738 in combination with gemcitabine merits evaluation in a clinical trial in patients with PDAC. Mol Cancer Ther; 17(8); 1670–82. ©2018 AACR.
- Published
- 2018
46. A phase I trial of the γ-secretase inhibitor MK-0752 in combination with gemcitabine in patients with pancreatic ductal adenocarcinoma
- Author
-
Duncan I. Jodrell, Daniel H. Palmer, Sarah Halford, J. Evans, Lisa V. Hampson, Balaji Venugopal, Robert McLeod, Mirela Hategan, Natalie Cook, Helen Turner, Donna Michelle Smith, Bristi Basu, Aarthi Gopinathan, Michael Nebozhyn, D. Alan Anthoney, David A. Tuveson, William P. Steward, David Propper, and Hayley Farmer-Hall
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,pancreatic cancer ,Notch pathway ,γ-secretase ,Deoxycytidine ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Benzene Derivatives ,Sulfones ,Infusions, Intravenous ,Receptors, Notch ,Manchester Cancer Research Centre ,gemcitabine ,Area under the curve ,Middle Aged ,G-secretase ,030220 oncology & carcinogenesis ,Female ,notch pathway ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,medicine.drug ,Adult ,medicine.medical_specialty ,Notch signaling pathway ,Drug Administration Schedule ,03 medical and health sciences ,Internal medicine ,Pancreatic cancer ,medicine ,Carcinoma ,Humans ,Aged ,Performance status ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Cancer ,Bayes Theorem ,medicine.disease ,Gemcitabine ,Pancreatic Neoplasms ,Clinical trial ,030104 developmental biology ,Clinical Study ,Amyloid Precursor Protein Secretases ,Propionates ,business - Abstract
Background: The Notch pathway is frequently activated in cancer. Pathway inhibition by g-secretase inhibitors has been shown to be effective in pre-clinical models of pancreatic cancer, in combination with gemcitabine. Methods: A multi-centre, non-randomised Bayesian adaptive design study of MK-0752, administered per os weekly, in combination with gemcitabine administered intravenously on days 1, 8 and 15 (28 day cycle) at 800 or 1000mgm-2, was performed to determine the safety of combination treatment and the recommended phase 2 dose (RP2D). Secondary and tertiary objectives included tumour response, plasma and tumour MK-0752 concentration, and inhibition of the Notch pathway in hair follicles and tumour. Results: Overall, 44 eligible patients (performance status 0 or 1 with adequate organ function) received gemcitabine and MK-0752 as first or second line treatment for pancreatic cancer. RP2Ds of MK-0752 and gemcitabine as single agents could be combined safely. The Bayesian algorithm allowed further dose escalation, but pharmacokinetic analysis showed no increase in MK-0752 AUC (area under the curve) beyond 1800mg once weekly. Tumour response evaluation was available in 19 patients; 13 achieved stable disease and 1 patient achieved a confirmed partial response. Conclusions: Gemcitabine and a g-secretase inhibitor (MK-0752) can be combined at their full, single-agent RP2Ds.
- Published
- 2018
47. Pancreatic Resectability in Cancers with Known Limited Extension (PRICKLE) – a single-arm phase 2a study of gemcitabine (Gem) plus Nab-paclitaxel (NabP) for borderline (un)resectable pancreatic ductal adenocarcinoma (BrPDAC)
- Author
-
Nicholas Carroll, Siong-Seng Liau, Richard Charnley, Siobhan Whitley, Rebecca Brais, Hemant M. Kocher, Amanda Walker, Ferdia A. Gallagher, Lisa Bax, Duncan I. Jodrell, Mary A. McLean, Edmund M. Godfrey, J. Scott, Wendi Qian, Bristi Basu, Andrea Machin, Aarthi Gopinathan, Joanna Calder, Colin D. Johnson, and Mark Duxbury
- Subjects
Pancreatic ductal adenocarcinoma ,Hepatology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Gastroenterology ,Cancer research ,medicine ,business ,Gemcitabine ,medicine.drug ,Nab-paclitaxel - Published
- 2020
48. Abstract CT214: ATRiUM: A first-in-human dose escalation phase I trial of ceralasertib (AZD6738) and gemcitabine as combination therapy
- Author
-
Purity Bundi, Emma Dean, Jeff Evans, Bruno Henrique Rala De Paula, James Thaventhiran, Richard J. A. Goodwin, Maria Pawula, Tatiana Hernandez, Subha Anand, Josephine Khan, Duncan I. Jodrell, Adrian Mander, Simon Smith, Gonzalez-Lopes Alberto, Eva M. Serrao, Sabita Islam, Vincenzo Graziano, Godfrey Edmund, and Bristi Basu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,business.industry ,Cancer ,medicine.disease ,Gemcitabine ,Pharmacokinetics ,Tolerability ,Pharmacodynamics ,Pancreatic cancer ,Internal medicine ,Toxicity ,medicine ,business ,medicine.drug - Abstract
Background:ATRiUM is a phase 1 trial to assess the safety, tolerability, pharmacokinetics and preliminary antitumour activity of ascending doses of combined therapy with the ATR inhibitor ceralasertib AZD6738 and gemcitabine, Using a Model based design NCT03669601. The combination was shown to act synergistically in laboratory models of pancreatic cancer. Based on these data, ATRiUM is now open to recruitment in Cambridge and Glasgow, UK. Methods:ATRiUM utilises a novel Bayesian model-based design for dual-agent dose escalation, that handles three levels of toxicity the highest of which is a dose limiting toxicity and two dosing schedules. The model assumes that the toxicity levels come from a multinomial distribution and toxicity increases monotonically as doses increase. Different schedules and doses 40-120 mg of ceralasertib; intermittent days 1-4, 8-11, 15-18 and continuous days 1-21 with gemcitabine 625-1000 mg/m2 administered on days 3, 10 and 17 of a 28-day cycle, will be assessed using the Bayesian framework. The aim is to recruit up to 55 patients, including up to 40 in the dose escalation phase across 3 gemcitabine doses and 6 ceralasertib dose/schedules. An expansion cohort (10-15 patients) is planned in patients with pancreatic cancer, based on our pre-clinical findings. Tumour monitoring will be undertaken by CT imaging RECIST 1.1, liquid biopsies circulating free DNA and advanced imaging radiomics. Baseline tissue biopsies will be analysed to identify potential predictive biomarkers e.g. ATM loss, as described pre-clinically. Imaging MSI will be used to characterise the intra-tumoural distribution of gemcitabine, its metabolites and ceralasertib and compared to plasma PK. Detailed studies of sequential tumour biopsies will include immunohistochemistry, RNAseq and Imaging Mass Cytometry IMC and be used to evaluate early pharmacodynamic responses to the combination. To investigate whether the combination increases tumour immunogenicity, the immune effects of the combination will be evaluated in both tumour by IMC and RNAseq and peripherally, by the serial collection and analysis of PBMCs and plasma for cytokine analysis. Adults with solid tumours are eligible for the dose escalation, if fulfilling standard Phase I eligibility criteria with measurable tumour lesions amenable to biopsy. Two out of six planned cohorts have been completed with no DLTs identified and cohort 3 will open in January 2021. ATRiUM is supported by the Integrative Cancer Medicine theme of the CRUK Cambridge Centre, the NIHR Cambridge Biomedical Centre, the Cambridge ECMC and a grant from AstraZeneca. Citation Format: Bruno Henrique de Paula, Bristi Basu, Adrian Mander, Josephine Khan, Purity Bundi, Richard Goodwin, James Thaventhiran, Gonzalez-Lopes Alberto, Godfrey Edmund, Sabita Islam, Subha Anand, Jeff Evans, Vincenzo Graziano, Eva Serrao, Maria Pawula, Tatiana Hernandez, Emma Dean, Simon Smith, Duncan Jodrell. ATRiUM: A first-in-human dose escalation phase I trial of ceralasertib (AZD6738) and gemcitabine as combination therapy [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 CT214.
- Published
- 2021
49. Abstract OT1-04-01: Cambridge brain mets trial 1 (CamBMT1): A proof-of-principle phase 1b / randomised phase 2 study of afatinib penetration into brain metastases for patients undergoing neurosurgical resection, both with and without prior low-dose, targeted radiotherapy
- Author
-
Stephen J. Price, Nicola Ramenatte, Louise Jones, Anthony J. Chalmers, A Jonson, Matthew Williams, L Dunn, Rajesh Jena, Wendi Qian, Fiona J. Gilbert, K Karabatsou, I Oberg, Colin Watts, Duncan I. Jodrell, G Bullen, Emma Harrison, Gillian A Whitfield, Heather Biggs, Richard D. Baird, Sarah Jefferies, and Carlos Caldas
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,business.industry ,Afatinib ,medicine.medical_treatment ,Phases of clinical research ,Drug resistance ,medicine.disease ,Surgery ,Radiation therapy ,medicine.anatomical_structure ,Breast cancer ,Statistical significance ,Internal medicine ,Clinical endpoint ,medicine ,business ,medicine.drug - Abstract
Background Failure of drugs to cross the blood brain barrier (BBB) can be a major reason for treatment failure for patients with brain tumors. For most patients who don't respond to treatment, it is not known whether this is due to inadequate drug concentrations in the tumor, or due to drug resistance. Preliminary data suggest that low-dose radiotherapy may disrupt the BBB, and could facilitate increased drug delivery into brain tumors. Afatinib is a potent, irreversible inhibitor of EGFR / HER2 / HER4 and takes approximately 8 days to achieve steady-state concentrations in cancer patients. Aims CamBMT1 has been designed to investigate the delivery of afatinib into brain metastases and whether this might be enhanced by low dose-radiotherapy. Patient Population Key eligibility criteria Patients with operable brain metastases from breast or lung primaries for whom neurosurgical resection would be standard of care, as determined by the local multi-disciplinary team. ECOG PS 0, 1 or 2. Trial design After a phase 1b safety run- in, the phase 2 part of the trial randomises patients (n=60) into 3 pre-operative arms: Arm 1afatinib alone for 11 days, then neurosurgery on day 12Arm 2afatinib for 11 days plus a single 2 Gy fraction on day 10, then neurosurgery on day 12Arm 3afatinib for 11 days plus a single 4 Gy fraction on day 10, then neurosurgery on day 12 Primary endpoint: to compare steady-state afatinib concentration in resected brain metastases, following afatinib administered alone, or in combination with radiotherapy (2 Gy or 4 Gy). Afatinib concentrations are measured in the resected brain metastases and in plasma. Secondary endpoints: safety of afatinib administration in combination with radiotherapy; and multi-sequence MRI (optional) to detect changes in perfusion, vascular density, blood-brain-barrier permeability and interstitial pressure. Exploratory endpoints: molecular profiling of resected brain metastases, for comparison with paired primary lung and breast cancers; the establishment and study of patient-derived xenografts. Statistical methods With 20 patients randomised in each of 3 arms in the phase 2 part of CamBMT1, the trial has a power of 84% at a significance level of 20% (one-sided) to detect an increase in afatinib concentrations with targeted radiotherapy, measured as a Cohen's D (standardised mean difference) ≥0.5. Accrual By the end of q2 2016, phase 1b had nearly completed enrolment. The randomised phase 2 part of CamBMT1 is due to open by q4 2016 at additional Experimental Cancer Medicine Centres. Acknowledgments CamBMT1 is funded by Cancer Research UK, the Brain Tumour Charity and Boehringer-Ingelheim. Citation Format: Baird RD, Ramenatte N, Watts C, Jonson A, Jones L, Biggs H, Harrison E, Oberg I, Bullen G, Williams M, Qian W, Gilbert F, Jodrell D, Caldas C, Karabatsou K, Dunn L, Jena R, Whitfield G, Chalmers A, Jefferies S, Price S. Cambridge brain mets trial 1 (CamBMT1): A proof-of-principle phase 1b / randomised phase 2 study of afatinib penetration into brain metastases for patients undergoing neurosurgical resection, both with and without prior low-dose, targeted radiotherapy [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr OT1-04-01.
- Published
- 2017
50. A Highly Porous Metal-Organic Framework System to Deliver Payloads for Gene Knockdown
- Author
-
Meng Lu, Peyman Z. Moghadam, Michelle H. Teplensky, David Fairen-Jimenez, Omar K. Farha, Chetan Poudel, Marcus Fantham, Clemens F. Kaminski, Sandra Bernaldo de Quirós Fernández, Alina Guna, Colin Hockings, Duncan I. Jodrell, Marta Aragones-Anglada, Frances M. Richards, Gabriele Kaminski Schierle, Peng Li, Poudel, Chetan [0000-0002-8512-9238], Hockings, Colin [0000-0002-0248-0517], Richards, Fran [0000-0001-7947-7853], Jodrell, Duncan [0000-0001-9360-1670], Kaminski Schierle, Gabriele [0000-0002-1843-2202], Kaminski, Clemens [0000-0002-5194-0962], Fairen Jimenez, David [0000-0002-5013-1194], and Apollo - University of Cambridge Repository
- Subjects
Small interfering RNA ,Endosome ,General Chemical Engineering ,Endocytic cycle ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Biochemistry ,RNA interference ,Materials Chemistry ,Genetics ,Environmental Chemistry ,Gene ,Gene knockdown ,34 Chemical Sciences ,Chemistry ,fungi ,Biochemistry (medical) ,RNA ,General Chemistry ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,Cell biology ,FOS: Biological sciences ,Drug delivery ,0210 nano-technology ,Biotechnology - Abstract
Summary Since first reported, RNA interference (RNAi) has become a widely used tool for cellular genetic knockdown. However, RNA instability and susceptibility to enzymatic degradation have prevented its widespread clinical use. Thus, research efforts are seeking methods to protect the fragile RNA payload during delivery. Here, we report the use of a metal-organic framework (MOF) to load, protect, and deliver small interfering ribonucleic acids (siRNAs). We confirmed the protection of MOF-internalized siRNA from enzymatic degradation. Furthermore, through combined encapsulation of siRNA in the MOF with various cofactors (proton sponge, KALA peptide, and NH4Cl), we show that endosomal retention can be evaded and ensure that gene knockdown is efficacious. In vitro studies after siRNA-MOF complexation demonstrated up to 27% consistent knockdown. We use structured illumination microscopy (SIM) to study the complex’s endocytic uptake. Overall, we demonstrate the potential of these highly porous and biodegradable materials to improve the efficacy and efficiency of future gene therapies.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.