194 results on '"Ovarian Cancer Action"'
Search Results
2. Polymorphic microbes: a new emerging hallmark of cancer
- Author
-
Mark P. Lythgoe, Benjamin H. Mullish, Adam E. Frampton, Jonathan Krell, Imperial College Healthcare NHS Trust- BRC Funding, 4D Pharma Plc, and Ovarian Cancer Action
- Subjects
Microbiology (medical) ,intratumoural microbiome ,polymorphic microbes ,Microbiota ,gut microbiome ,Microbiology ,Gastrointestinal Microbiome ,Infectious Diseases ,1108 Medical Microbiology ,Neoplasms ,Virology ,live biotherapeutic ,Humans ,cancer hallmarks ,faecal microbiota transplantation ,0605 Microbiology - Abstract
Recognition of the microbiome (and 'polymorphic microbes' within them) as a new emerging hallmark of cancer reflects a wide body of rapidly evolving research. Microbes may be directly carcinogenic, impact host immune responses to promote malignancy, and may be key effectors in determining the efficacy of anticancer therapy. Manipulation of the microbiome is showing promise as an opportunity to influence cancer outcomes.
- Published
- 2022
3. Weekly dose-dense chemotherapy in first-line epithelial ovarian, fallopian tube, or primary peritoneal cancer treatment (ICON8): overall survival results from an open-label, randomised, controlled, phase 3 trial
- Author
-
Andrew R Clamp, Elizabeth C James, Iain A McNeish, Andrew Dean, Jae-Won Kim, Dearbhaile M O'Donnell, Dolores Gallardo-Rincon, Sarah Blagden, James Brenton, Tim J Perren, Sudha Sundar, Rosemary Lord, Graham Dark, Marcia Hall, Susana Banerjee, Rosalind M Glasspool, C Louise Hanna, Sarah Williams, Kate M Scatchard, Helena Nam, Sharadah Essapen, Christine Parkinson, Lucy McAvan, Ann Marie Swart, Babasola Popoola, Francesca Schiavone, Jonathan Badrock, Fuad Fananapazir, Adrian D Cook, Mahesh Parmar, Richard Kaplan, Jonathan A Ledermann, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Ovarian Neoplasms ,Paclitaxel ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,1112 Oncology and Carcinogenesis ,Antineoplastic Agents ,Female ,Oncology & Carcinogenesis ,Carcinoma, Ovarian Epithelial ,Middle Aged ,Fallopian Tubes ,Carboplatin - Abstract
BACKGROUND: Standard-of-care first-line chemotherapy for epithelial ovarian cancer is carboplatin and paclitaxel administered once every 3 weeks. The JGOG 3016 trial reported significant improvement in progression-free and overall survival with dose-dense weekly paclitaxel and 3-weekly (ie, once every 3 weeks) carboplatin. However, this benefit was not observed in the previously reported progression-free survival results of ICON8. Here, we present the final coprimary outcomes of overall survival and updated progression-free survival analyses of ICON8. METHODS: In this open-label, randomised, controlled, phase 3 trial (ICON8), women aged 18 years or older with newly diagnosed stage IC-IV epithelial ovarian, primary peritoneal, or fallopian tube carcinoma (here collectively termed ovarian cancer, as defined by International Federation of Gynecology and Obstetrics [FIGO] 1988 criteria) and an Eastern Cooperative Oncology Group performance status of 0-2 were recruited from 117 hospitals with oncology departments in the UK, Australia and New Zealand, Mexico, South Korea, and Ireland. Patients could enter the trial after immediate primary surgery (IPS) or with planned delayed primary surgery (DPS) during chemotherapy, or could have no planned surgery. Participants were randomly assigned (1:1:1), using the Medical Research Council Clinical Trials Unit at University College London randomisation line with stratification by Gynecologic Cancer Intergroup group, FIGO disease stage, and outcome and timing of surgery, to either 3-weekly carboplatin area under the curve (AUC)5 or AUC6 and 3-weekly paclitaxel 175 mg/m2 (control; group 1), 3-weekly carboplatin AUC5 or AUC6 and weekly paclitaxel 80 mg/m2 (group 2), or weekly carboplatin AUC2 and weekly paclitaxel 80 mg/m2 (group 3), all administered via intravenous infusion for a total of six 21-day cycles. Coprimary outcomes were progression-free survival and overall survival, with comparisons done between group 2 and group 1, and group 3 and group 1, in the intention-to-treat population. Safety was assessed in all patients who started at least one chemotherapy cycle. The trial is registered on ClinicalTrials.gov, NCT01654146, and ISRCTN registry, ISRCTN10356387, and is closed to accrual. FINDINGS: Between June 6, 2011, and Nov 28, 2014, 1566 patients were randomly assigned to group 1 (n=522), group 2 (n=523), or group 3 (n=521). The median age was 62 years (IQR 54-68), 1073 (69%) of 1566 patients had high-grade serous carcinoma, 1119 (71%) had stage IIIC-IV disease, and 745 (48%) had IPS. As of data cutoff (March 31, 2020), with a median follow-up of 69 months (IQR 61-75), no significant difference in overall survival was observed in either comparison: median overall survival of 47·4 months (95% CI 43·1-54·8) in group 1, 54·8 months (46·6-61·6) in group 2, and 53·4 months (49·2-59·6) in group 3 (group 2 vs group 1: hazard ratio 0·87 [97·5% CI 0·73-1·05]; group 3 vs group 1: 0·91 [0·76-1·09]). No significant difference was observed for progression-free survival in either comparison and evidence of non-proportional hazards was seen (p=0·037), with restricted mean survival time of 23·9 months (97·5% CI 22·1-25·6) in group 1, 25·3 months (23·6-27·1) in group 2, and 24·8 months (23·0-26·5) in group 3. The most common grade 3-4 adverse events were reduced neutrophil count (78 [15%] of 511 patients in group 1, 183 [36%] of 514 in group 2, and 154 [30%] of 513 in group 3), reduced white blood cell count (22 [4%] in group 1, 80 [16%] in group 2, and 71 [14%] in group 3), and anaemia (26 [5%] in group 1, 66 [13%] in group 2, and 24 [5%] in group 3). No new serious adverse events were reported. Seven treatment-related deaths were reported (two in group 1, four in group 2, and one in group 3). INTERPRETATION: In our cohort of predominantly European women with epithelial ovarian cancer, we found that first-line weekly dose-dense chemotherapy did not improve overall or progression-free survival compared with standard 3-weekly chemotherapy and should not be used as part of standard multimodality front-line therapy in this patient group. FUNDING: Cancer Research UK, Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, and Cancer Australia.
- Published
- 2022
4. Investigation of the Thermogelation of a Promising Biocompatible ABC Triblock Terpolymer and Its Comparison with Pluronic F127
- Author
-
Anna P. Constantinou, Valeria Nele, James J. Doutch, Joana S. Correia, Roman V. Moiseev, Martina Cihova, David C. A. Gaboriau, Jonathan Krell, Vitaliy V. Khutoryanskiy, Molly M. Stevens, Theoni K. Georgiou, Engineering & Physical Science Research Council (E, ISIS Neutron & Muon Source, Engineering and Physical Sciences Research Council, and Ovarian Cancer Action
- Subjects
Science & Technology ,Polymers and Plastics ,Polymers ,TRANSFECTION EFFICIENCY ,Organic Chemistry ,Polymer Science ,2-(2-METHOXYETHOXY)ETHYL METHACRYLATE ,HIGHLY EFFICIENT ,09 Engineering ,GROUP-TRANSFER POLYMERIZATION ,Inorganic Chemistry ,BLOCK-COPOLYMERS ,MOLECULAR-WEIGHT ,NEUTRON-SCATTERING ,Physical Sciences ,AQUEOUS-SOLUTIONS ,THERMORESPONSIVE HYDROGELS ,Materials Chemistry ,PHASE-TRANSITIONS ,03 Chemical Sciences - Abstract
Thermoresponsive polymers with the appropriate structure form physical networks upon changes in temperature, and they find utility in formulation science, tissue engineering, and drug delivery. Here, we report a cost-effective biocompatible alternative, namely OEGMA30015-b-BuMA26-b-DEGMA13, which forms gels at low concentrations (as low as 2% w/w); OEGMA300, BuMA, and DEGMA stand for oligo(ethylene glycol) methyl ether methacrylate (MM = 300 g mol–1), n-butyl methacrylate, and di(ethylene glycol) methyl ether methacrylate, respectively. This polymer is investigated in depth and is compared to its commercially available counterpart, Poloxamer P407 (Pluronic F127). To elucidate the differences in their macroscale gelling behavior, we investigate their nanoscale self-assembly by means of small-angle neutron scattering and simultaneously recording their rheological properties. Two different gelation mechanisms are revealed. The triblock copolymer inherently forms elongated micelles, whose length increases by temperature to form worm-like micelles, thus promoting gelation. In contrast, Pluronic F127’s micellization is temperature-driven, and its gelation is attributed to the close packing of the micelles. The gel structure is analyzed through cryogenic scanning and transmission electron microscopy. Ex vivo gelation study upon intracameral injections demonstrates excellent potential for its application to improve drug residence in the eye.
- Published
- 2022
5. Dual G9A/EZH2 Inhibition Stimulates Antitumor Immune Response in Ovarian High-Grade Serous Carcinoma
- Author
-
Pavlina Spiliopoulou, Sarah Spear, Hasan Mirza, Ian Garner, Lynn McGarry, Fabio Grundland-Freile, Zhao Cheng, Darren P. Ennis, Nayana Iyer, Sophie McNamara, Marina Natoli, Susan Mason, Karen Blyth, Peter D. Adams, Patricia Roxburgh, Matthew J. Fuchter, Bob Brown, Iain A. McNeish, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, National Institute for Health Research, and Engineering & Physical Science Research Council (EPSRC)
- Subjects
Ovarian Neoplasms ,Cancer Research ,Immunity ,Carcinoma, Ovarian Epithelial ,Prognosis ,Cystadenocarcinoma, Serous ,Epigenesis, Genetic ,Mice ,Oncology ,Tumor Microenvironment ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,1115 Pharmacology and Pharmaceutical Sciences - Abstract
Ovarian high-grade serous carcinoma (HGSC) prognosis correlates directly with presence of intratumoral lymphocytes. However, cancer immunotherapy has yet to achieve meaningful survival benefit in patients with HGSC. Epigenetic silencing of immunostimulatory genes is implicated in immune evasion in HGSC and re-expression of these genes could promote tumor immune clearance. We discovered that simultaneous inhibition of the histone methyltransferases G9A and EZH2 activates the CXCL10–CXCR3 axis and increases homing of intratumoral effector lymphocytes and natural killer cells while suppressing tumor-promoting FoxP3+ CD4 T cells. The dual G9A/EZH2 inhibitor HKMTI-1–005 induced chromatin changes that resulted in the transcriptional activation of immunostimulatory gene networks, including the re-expression of elements of the ERV-K endogenous retroviral family. Importantly, treatment with HKMTI-1–005 improved the survival of mice bearing Trp53−/− null ID8 ovarian tumors and resulted in tumor burden reduction. These results indicate that inhibiting G9A and EZH2 in ovarian cancer alters the immune microenvironment and reduces tumor growth and therefore positions dual inhibition of G9A/EZH2 as a strategy for clinical development.
- Published
- 2022
6. Characterization of patients with long-term responses to rucaparib treatment in recurrent ovarian cancer
- Author
-
Carol Aghajanian, Lee-may Chen, Scott H. Kaufmann, Amit M. Oza, Geoffrey I. Shapiro, Rebecca Kristeleit, Sandra Goble, Iain A. McNeish, Diane Provencher, Ana Oaknin, Alexandra Leary, Lara Maloney, Elizabeth M. Swisher, Isabelle Ray-Coquard, Howard A. Burris, Stephen Welch, Kevin K. Lin, Tanya Kwan, Ronnie Shapira-Frommer, Anna V. Tinker, David M. O'Malley, Robert L. Coleman, Cancer Research UK, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Oncology ,Indoles ,ANTITUMOR-ACTIVITY ,MONOTHERAPY ,Loss of Heterozygosity ,DOUBLE-BLIND ,chemistry.chemical_compound ,Ovarian carcinoma ,Aged, 80 and over ,Ovarian Neoplasms ,Clinical Trials, Phase I as Topic ,BRCA1 Protein ,OLAPARIB ,Obstetrics & Gynecology ,Obstetrics and Gynecology ,Structural variant ,Genomics ,Middle Aged ,PARP inhibitor ,SURVIVAL ,RAD51C ,Female ,Safety ,Life Sciences & Biomedicine ,Adult ,medicine.medical_specialty ,CARCINOMA ,Duration of response ,Poly(ADP-ribose) Polymerase Inhibitors ,Clinical Trials, Phase II as Topic ,Internal medicine ,GERMLINE ,Post-hoc analysis ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Rucaparib ,Aged ,BRCA2 Protein ,Science & Technology ,MUTATIONS ,business.industry ,BRCA1 ,chemistry ,PARP INHIBITOR RUCAPARIB ,Recurrent Ovarian Cancer ,1114 Paediatrics and Reproductive Medicine ,Neoplasm Recurrence, Local ,business ,Recurrent Ovarian Carcinoma ,Follow-Up Studies - Abstract
Objective. To describe molecular and clinical characteristics of patients with high-grade recurrent ovarian carcinoma (HGOC) who had long-term responses to the poly(ADP-ribose) polymerase (PARP) inhibitor rucaparib. Methods. This post hoc analysis pooled patients from Study 10 (NCT01482715; Parts 2A and 2B; n = 54) and ARIEL2 (NCT01891344; Parts 1 and 2; n = 491). Patients with investigator-assessed complete or partial response per RECIST were classified based on duration of response (DOR): long (≥1 year), intermediate (6 months to
- Published
- 2021
7. A Randomized, Phase III Trial to Evaluate Rucaparib Monotherapy as Maintenance Treatment in Patients With Newly Diagnosed Ovarian Cancer (ATHENA–MONO/GOG-3020/ENGOT-ov45)
- Author
-
Bradley J. Monk, Christine Parkinson, Myong Cheol Lim, David M. O'Malley, Ana Oaknin, Michelle K. Wilson, Robert L. Coleman, Domenica Lorusso, Paul Bessette, Sharad Ghamande, Athina Christopoulou, Diane Provencher, Emily Prendergast, Fuat Demirkiran, Olga Mikheeva, Oladapo Yeku, Anita Chudecka-Glaz, Michael Schenker, Ramey D. Littell, Tamar Safra, Hung-Hsueh Chou, Mark A. Morgan, Vít Drochýtek, Joyce N. Barlin, Toon Van Gorp, Fred Ueland, Gabriel Lindahl, Charles Anderson, Dearbhaile C. Collins, Kathleen Moore, Frederik Marme, Shannon N. Westin, Iain A. McNeish, Danny Shih, Kevin K. Lin, Sandra Goble, Stephanie Hume, Keiichi Fujiwara, Rebecca S. Kristeleit, Institut Català de la Salut, [Monk BJ] GOG Foundation, HonorHealth Research Institute, University of Arizona College of Medicine, Creighton University School of Medicine, Phoenix, AZ. [Parkinson C] Medical Oncology, Addenbrooke's Hospital, Cambridge, United Kingdom. [Lim MC] Gynecologic Oncology, National Cancer Center Korea, Goyang-si, Gyeonggi-do, South Korea. [O'Malley DM] Division of Gynecologic Oncology, The Ohio State University, James Cancer Center, Columbus, OH. [Oaknin A] Gynaecologic Cancer Programme, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Vall d'Hebron Hospital Universitari, Barcelona, Spain. [Wilson MK] Department of Cancer and Blood, Auckland City Hospital, Auckland, New Zealand, Vall d'Hebron Barcelona Hospital Campus, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Cancer Research ,Indoles ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Poly(ADP-ribose) Polymerase Inhibitors ,Carcinoma, Ovarian Epithelial ,neoplasias::neoplasias por localización::neoplasias de las glándulas endocrinas::neoplasias ováricas [ENFERMEDADES] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Maintenance Chemotherapy ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Other subheadings::/therapeutic use [Other subheadings] ,Ovarian Neoplasms ,Cancer och onkologi ,Otros calificadores::/uso terapéutico [Otros calificadores] ,Neoplasms::Neoplasms by Site::Endocrine Gland Neoplasms::Ovarian Neoplasms [DISEASES] ,diagnóstico::pronóstico::resultado del tratamiento [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,1103 Clinical Sciences ,Ovaris - Càncer - Tractament ,Diagnosis::Prognosis::Treatment Outcome [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Oncology ,Cancer and Oncology ,Avaluació de resultats (Assistència sanitària) ,Female ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] - Abstract
PURPOSE ATHENA (ClinicalTrials.gov identifier: NCT03522246 ) was designed to evaluate rucaparib first-line maintenance treatment in a broad patient population, including those without BRCA1 or BRCA2 (BRCA) mutations or other evidence of homologous recombination deficiency (HRD), or high-risk clinical characteristics such as residual disease. We report the results from the ATHENA–MONO comparison of rucaparib versus placebo. METHODS Patients with stage III-IV high-grade ovarian cancer undergoing surgical cytoreduction (R0/complete resection permitted) and responding to first-line platinum-doublet chemotherapy were randomly assigned 4:1 to oral rucaparib 600 mg twice a day or placebo. Stratification factors were HRD test status, residual disease after chemotherapy, and timing of surgery. The primary end point of investigator-assessed progression-free survival was assessed in a step-down procedure, first in the HRD population (BRCA-mutant or BRCA wild-type/loss of heterozygosity high tumor), and then in the intent-to-treat population. RESULTS As of March 23, 2022 (data cutoff), 427 and 111 patients were randomly assigned to rucaparib or placebo, respectively (HRD population: 185 v 49). Median progression-free survival (95% CI) was 28.7 months (23.0 to not reached) with rucaparib versus 11.3 months (9.1 to 22.1) with placebo in the HRD population (log-rank P = .0004; hazard ratio [HR], 0.47; 95% CI, 0.31 to 0.72); 20.2 months (15.2 to 24.7) versus 9.2 months (8.3 to 12.2) in the intent-to-treat population (log-rank P < .0001; HR, 0.52; 95% CI, 0.40 to 0.68); and 12.1 months (11.1 to 17.7) versus 9.1 months (4.0 to 12.2) in the HRD-negative population (HR, 0.65; 95% CI, 0.45 to 0.95). The most common grade ≥ 3 treatment-emergent adverse events were anemia (rucaparib, 28.7% v placebo, 0%) and neutropenia (14.6% v 0.9%). CONCLUSION Rucaparib monotherapy is effective as first-line maintenance, conferring significant benefit versus placebo in patients with advanced ovarian cancer with and without HRD.
- Published
- 2022
8. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer
- Author
-
Martins, Filipe Correia, Couturier, Dominique-Laurent, De Santiago, Ines, Sauer, Carolin Margarethe, Vias, Maria, Angelova, Mihaela, Sanders, Deborah, Piskorz, Anna, Hall, James, Hosking, Karen, Amirthanayagam, Anumithra, Cosulich, Sabina, Carnevalli, Larissa, Davies, Barry, Watkins, Thomas BK, Funingana, Ionut G, Bolton, Helen, Haldar, Krishnayan, Latimer, John, Baldwin, Peter, Crawford, Robin, Eldridge, Matthew, Basu, Bristi, Jimenez-Linan, Mercedes, Mcpherson, Andrew W, McGranahan, Nicholas, Litchfield, Kevin, Shah, Sohrab P, McNeish, Iain, Caldas, Carlos, Evan, Gerard, Swanton, Charles, Brenton, James D, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, National Institute for Health Research, Cancer Research UK, Martins, Filipe Correia [0000-0001-6459-8206], Couturier, Dominique-Laurent [0000-0001-5774-5036], de Santiago, Ines [0000-0002-8172-1322], Sauer, Carolin Margarethe [0000-0003-2168-6630], Angelova, Mihaela [0000-0002-0495-9695], Carnevalli, Larissa [0000-0001-7432-0195], Davies, Barry [0000-0002-5414-9113], Funingana, Ionut G [0000-0002-1197-2652], Eldridge, Matthew [0000-0002-5799-8911], Basu, Bristi [0000-0002-3562-2868], Litchfield, Kevin [0000-0002-3725-0914], Shah, Sohrab P [0000-0001-6402-523X], McNeish, Iain [0000-0002-9387-7586], Caldas, Carlos [0000-0003-3547-1489], Evan, Gerard [0000-0003-0412-1216], Swanton, Charles [0000-0002-4299-3018], Brenton, James D [0000-0002-5738-6683], and Apollo - University of Cambridge Repository
- Subjects
DNA Copy Number Variations ,Paclitaxel ,Class I Phosphatidylinositol 3-Kinases ,FORMALIN ,General Physics and Astronomy ,PROTEIN ,631/67/69 ,Mechanistic Target of Rapamycin Complex 1 ,13 ,General Biochemistry, Genetics and Molecular Biology ,38 ,SYNTHETIC LETHALITY ,Proto-Oncogene Proteins p21(ras) ,Signalling & Oncogenes ,Phosphatidylinositol 3-Kinases ,Ecology,Evolution & Ethology ,C-MYC ,Humans ,SIGNATURES ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Ovarian Neoplasms ,Human Biology & Physiology ,Multidisciplinary ,Science & Technology ,45 ,LANDSCAPE ,631/67/1517/1709 ,TOR Serine-Threonine Kinases ,Genome Integrity & Repair ,article ,PATHWAYS ,PLATFORM ,General Chemistry ,DNA ,Tumour Biology ,692/4028/67 ,REGIONS ,49 ,Cystadenocarcinoma, Serous ,Multidisciplinary Sciences ,631/67/1059/602 ,Science & Technology - Other Topics ,Female ,631/67/70 ,Genetics & Genomics - Abstract
Chromosomal instability is a major challenge to patient stratification and targeted drug development for high-grade serous ovarian carcinoma (HGSOC). Here we show that somatic copy number alterations (SCNAs) in frequently amplified HGSOC cancer genes significantly correlate with gene expression and methylation status. We identify five prevalent clonal driver SCNAs (chromosomal amplifications encompassing MYC, PIK3CA, CCNE1, KRAS and TERT) from multi-regional HGSOC data and reason that their strong selection should prioritise them as key biomarkers for targeted therapies. We use primary HGSOC spheroid models to test interactions between in vitro targeted therapy and SCNAs. MYC chromosomal copy number is associated with in-vitro and clinical response to paclitaxel and in-vitro response to mTORC1/2 inhibition. Activation of the mTOR survival pathway in the context of MYC-amplified HGSOC is statistically associated with increased prevalence of SCNAs in genes from the PI3K pathway. Co-occurrence of amplifications in MYC and genes from the PI3K pathway is independently observed in squamous lung cancer and triple negative breast cancer. In this work, we show that identifying co-occurrence of clonal driver SCNA genes could be used to tailor therapeutics for precision medicine., F.C.M. is funded by the Experimental Medicine Initiative from the University of Cambridge, by the Academy of Medical Sciences (SGL016_1084), Cancer Research UK (C53876/A24267) and by the Addenbrooke’s Charitable Trust (REF 13/17). This research was also supported by a pump-priming award from the Cancer Research UK Cambridge Centre Early detection Programme (CRUK grant ref: A25117). This research was supported by the NIHR Cambridge Biomedical Research Centre. The OVO4 study is supported by the CRUK Cambridge Cancer Centre and the Mark Foundation Institute for Integrated Cancer Medicine. We would like to acknowledge the support of The University of Cambridge, the National Institute for Health Research Cambridge, National Cancer Research Network, the Cambridge Experimental Cancer Medicine Centres, Hutchison Whampoa Limited and Cancer Research UK (CRUK grant numbers A22905 (JDB), A15601 (JDB), A25177 (CRUK Cancer Centre Cambridge)). C.S. is Royal Society Napier Research Professor. His team and work is supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (FC001169), the UK Medical Research Council (FC001169), and the Wellcome Trust (FC001169). C.S. is funded by Cancer Research UK (TRACERx, PEACE and CRUK Cancer Immunotherapy Catalyst Network), Cancer Research UK Lung Cancer Centre of Excellence, the Rosetrees Trust, Butterfield and Stoneygate Trusts, NovoNordisk Foundation (ID16584), Royal Society Professorship Enhancement Award (RP/EA/180007), the National Institute for Health Research (NIHR) Biomedical Research Centre at University College London Hospitals, the CRUK-UCL Centre, Experimental Cancer Medicine Centre, and the Breast Cancer Research Foundation (BCRF, USA). His research is supported by a Stand Up To Cancer‐LUNGevity-American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT23-17). Stand Up To Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the Scientific Partner of SU2C. CS receives funding from the European Research Council (ERC) under the European Union’s Seventh Framework Programme (FP7/2007-2013) Consolidator Grant (FP7-THESEUS-617844), European Commission ITN (FP7-PloidyNet 607722), an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 835297), and Chromavision from the European Union’s Horizon 2020 research and innovation programme (grant agreement 665233).
- Published
- 2022
- Full Text
- View/download PDF
9. Methylation-based markers of aging and lifestyle-related factors and risk of breast cancer: a pooled analysis of four prospective studies
- Author
-
Pierre-Antoine Dugué, Clara Bodelon, Felicia F. Chung, Hannah R. Brewer, Srikant Ambatipudi, Joshua N. Sampson, Cyrille Cuenin, Veronique Chajès, Isabelle Romieu, Giovanni Fiorito, Carlotta Sacerdote, Vittorio Krogh, Salvatore Panico, Rosario Tumino, Paolo Vineis, Silvia Polidoro, Laura Baglietto, Dallas English, Gianluca Severi, Graham G. Giles, Roger L. Milne, Zdenko Herceg, Montserrat Garcia-Closas, James M. Flanagan, Melissa C. Southey, Monash University [Clayton], National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), Centre de recherche en épidémiologie et santé des populations (CESP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, National Institutes of Health, NIH, National Cancer Institute, NCI, Breast Cancer Now, BCN, Division of Cancer Epidemiology and Genetics, National Cancer Institute, DCEG, European Commission, EC, National Health and Medical Research Council, NHMRC: 1011618, 1164455, Fondation ARC pour la Recherche sur le Cancer, ARC, Ligue Contre le Cancer, Seventh Framework Programme, FP7, Institut National Du Cancer, INCa, Where authors are identified as personnel of the International Agency for Research on Cancer/World Health Organization, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/World Health Organization., This research was partially supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Division of Cancer Epidemiology and Genetics. The work performed by the Epigenomics and Mechanisms Branch at IARC was supported by grants from the Institut National du Cancer (INCa, France), the European Commission (EC) Seventh Framework Programme (FP7) Translational Cancer Research (TRANSCAN) Framework, the Fondation ARC pour la Recherche sur le Cancer (France), and la Ligue Nationale (Française) Contre le Cancer to ZH. The MCCS methylation work was supported by the National Health and Medical Research Council (grant numbers 1011618 and 1164455) and the Victorian Breast Cancer Research Consortium. The work performed at Imperial College London was funded by Breast Cancer Now and supported by the Cancer Research UK Imperial Centre, the National Institute for Health Research Imperial Biomedical Research Centre and the Ovarian Cancer Action Research Centre. MCS is a National Health and Medical Research Council Senior Research Fellow (GNT1155163)., and HAL UVSQ, Équipe
- Subjects
Aging ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Breast Neoplasms ,DNA Methylation ,Lifestyle ,Epigenesis, Genetic ,Epigenetic aging ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Risk Factors ,Prospective study DNA methylation Epigenetic aging Lifestyle Breast cancer risk ,Humans ,Female ,Prospective Studies ,Prospective study ,Breast cancer risk ,Life Style - Abstract
Background DNA methylation in blood may reflect adverse exposures accumulated over the lifetime and could therefore provide potential improvements in the prediction of cancer risk. A substantial body of research has shown associations between epigenetic aging and risk of disease, including cancer. Here we aimed to study epigenetic measures of aging and lifestyle-related factors in association with risk of breast cancer. Methods Using data from four prospective case–control studies nested in three cohorts of European ancestry participants, including a total of 1,655 breast cancer cases, we calculated three methylation-based measures of lifestyle factors (body mass index [BMI], tobacco smoking and alcohol consumption) and seven measures of epigenetic aging (Horvath-based, Hannum-based, PhenoAge and GrimAge). All measures were regression-adjusted for their respective risk factors and expressed per standard deviation (SD). Odds ratios (OR) and 95% confidence intervals (CI) were calculated using conditional or unconditional logistic regression and pooled using fixed-effects meta-analysis. Subgroup analyses were conducted by age at blood draw, time from blood sample to diagnosis, oestrogen receptor-positivity status and tumour stage. Results None of the measures of epigenetic aging were associated with risk of breast cancer in the pooled analysis: Horvath ‘age acceleration’ (AA): OR per SD = 1.02, 95%CI: 0.95–1.10; AA-Hannum: OR = 1.03, 95%CI:0.95–1.12; PhenoAge: OR = 1.01, 95%CI: 0.94–1.09 and GrimAge: OR = 1.03, 95%CI: 0.94–1.12, in models adjusting for white blood cell proportions, body mass index, smoking and alcohol consumption. The BMI-adjusted predictor of BMI was associated with breast cancer risk, OR per SD = 1.09, 95%CI: 1.01–1.17. The results for the alcohol and smoking methylation-based predictors were consistent with a null association. Risk did not appear to substantially vary by age at blood draw, time to diagnosis or tumour characteristics. Conclusion We found no evidence that methylation-based measures of aging, smoking or alcohol consumption were associated with risk of breast cancer. A methylation-based marker of BMI was associated with risk and may provide insights into the underlying associations between BMI and breast cancer.
- Published
- 2022
10. Polymeric carriers for delivery of RNA cancer therapeutics
- Author
-
Sofía Mirón-Barroso, Joana Correia, Adam Frampton, Mark Lythgoe, James Clark, Laura Tookman, Silvia Ottaviani, Leandro Castellano, Alexandra Porter, Theoni Georgiou, Jonathan Krell, and Ovarian Cancer Action
- Subjects
Genetics ,Molecular Biology ,Biochemistry - Abstract
As research uncovers the underpinnings of cancer biology, new targeted therapies have been developed. Many of these therapies are small molecules, such as kinase inhibitors, that target specific proteins; however, only 1% of the genome encodes for proteins and only a subset of these proteins has ‘druggable’ active binding sites. In recent decades, RNA therapeutics have gained popularity due to their ability to affect targets that small molecules cannot. Additionally, they can be manufactured more rapidly and cost-effectively than small molecules or recombinant proteins. RNA therapeutics can be synthesised chemically and altered quickly, which can enable a more personalised approach to cancer treatment. Even though a wide range of RNA therapeutics are being developed for various indications in the oncology setting, none has reached the clinic to date. One of the main reasons for this is attributed to the lack of safe and effective delivery systems for this type of therapeutic. This review focuses on current strategies to overcome these challenges and enable the clinical utility of these novel therapeutic agents in the cancer clinic.
- Published
- 2022
11. A novel cell line panel reveals non-genetic mediators of platinum resistance and phenotypic diversity in high grade serous ovarian cancer
- Author
-
J I, Hoare, H, Hockings, J, Saxena, V L, Silva, M J, Haughey, G E, Wood, F, Nicolini, H, Mirza, I A, McNeish, W, Huang, E, Maniati, T A, Graham, M, Lockley, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Ovarian Neoplasms ,Non-genetic ,Evolution ,Obstetrics and Gynecology ,Extracellular matrix ,Carcinoma, Ovarian Epithelial ,High grade serous ovarian cancer ,Carboplatin ,Cell Line ,Oncology ,Drug Resistance, Neoplasm ,Drug resistance ,Tumor Microenvironment ,Humans ,Chemotherapy ,1114 Paediatrics and Reproductive Medicine ,Female ,1112 Oncology and Carcinogenesis ,Gene expression ,Oncology & Carcinogenesis ,Cisplatin ,Transcription ,Platinum - Abstract
Objectives: Resistance to cancer therapy is an enduring challenge and accurate and reliable preclinical models are lacking. We interrogated this unmet need using high grade serous ovarian cancer (HGSC) as a disease model. Methods: We created five in vitro and two in vivo platinum-resistant HGSC models and characterised the entire cell panel via whole genome sequencing, RNASeq and creation of intraperitoneal models. Results: Mutational signature analysis indicated that platinum-resistant cell lines evolved from a pre existing ancestral clone but a unifying mutational cause for drug resistance was not identified. However, cisplatin-resistant and carboplatin-resistant cells evolved recurrent changes in gene expression that significantly overlapped with independent samples obtained from multiple patients with relapsed HGSC. Gene Ontology Biological Pathways (GOBP) related to the tumour microenvironment, particularly the extracellular matrix, were repeatedly enriched in cisplatin50 resistant cells, carboplatin-resistant cells and also in human resistant/refractory samples. The majority of significantly over-represented GOBP however, evolved uniquely in either cisplatin- or carboplatin52 resistant cell lines resulting in diverse intraperitoneal behaviours that reflect different clinical manifestations of relapsed human HGSC. Conclusions: Our clinically relevant and usable models reveal a key role for non-genetic factors in the evolution of chemotherapy resistance. Biological pathways relevant to the extracellular matrix were repeatedly expressed by resistant cancer cells in multiple settings. This suggests that recurrent gene expression changes provide a fitness advantage during platinum therapy and also that cancer cell intrinsic mechanisms influence the tumour microenvironment during the evolution of drug resistance. Candidate genes and pathways identified here could reveal therapeutic opportunities in platinum60 resistant HGSC.
- Published
- 2022
12. Response to the letter entitled 'Glomerular filtration rate estimation for carboplatin dosing in patients with gynaecological cancers'
- Author
-
A. Samani, J. Krell, I. McNeish, L. Tookman, and Ovarian Cancer Action
- Subjects
Cancer Research ,Science & Technology ,Oncology ,Genital Neoplasms, Female ,Humans ,Antineoplastic Agents ,Female ,CHEMOTHERAPY ,OVARIAN ,Life Sciences & Biomedicine ,CREATININE ,Carboplatin ,Glomerular Filtration Rate - Published
- 2022
13. Deciphering the role of histone modifications in uterine leiomyoma: acetylation of H3K27 regulates the expression of genes involved in proliferation, cell signaling, cell transport, angiogenesis and extracellular matrix formation
- Author
-
María Cristina Carbajo-García, Lucia de Miguel-Gómez, Elena Juárez-Barber, Alexandra Trelis, Javier Monleón, Antonio Pellicer, James M. Flanagan, Hortensia Ferrero, and Ovarian Cancer Action
- Subjects
Biochemistry & Molecular Biology ,Science & Technology ,uterine leiomyoma ,extracellular matrix ,PATHOGENESIS ,INVASION ,histone modification ,gene expression ,angiogenesis ,Medicine (miscellaneous) ,PATHWAYS ,Research & Experimental Medicine ,DIAGNOSIS ,CANCER ,General Biochemistry, Genetics and Molecular Biology ,SUBTYPES ,ACTIVATION ,Medicine, Research & Experimental ,POOR-PROGNOSIS ,SUPPRESSES ,Pharmacology & Pharmacy ,OVEREXPRESSION ,Life Sciences & Biomedicine - Abstract
Uterine leiomyoma (UL) is a benign tumor arising from myometrium (MM) with a high prevalence and unclear pathology. Histone modifications are altered in tumors, particularly via histone acetylation which is correlated with gene activation. To identify if the acetylation of H3K27 is involved in UL pathogenesis and if its reversion may be a therapeutic option, we performed a prospective study integrating RNA-seq (n = 48) and CHIP-seq for H3K27ac (n = 19) in UL vs MM tissue, together with qRT-PCR of SAHA-treated UL cells (n = 10). CHIP-seq showed lower levels of H3K27ac in UL versus MM (p-value < 2.2 × 10−16). From 922 DEGs found in UL vs. MM (FDR < 0.01), 482 presented H3K27ac. A differential acetylation (FDR < 0.05) was discovered in 82 of these genes (29 hyperacetylated/upregulated, 53 hypoacetylated/downregulated). Hyperacetylation/upregulation of oncogenes (NDP,HOXA13,COL24A1,IGFL3) and hypoacetylation/downregulation of tumor suppressor genes (CD40,GIMAP8,IL15,GPX3,DPT) altered the immune system, the metabolism, TGFβ3 and the Wnt/β-catenin pathway. Functional enrichment analysis revealed deregulation of proliferation, cell signaling, transport, angiogenesis and extracellular matrix. Inhibition of histone deacetylases by SAHA increased expression of hypoacetylated/downregulated genes in UL cells (p < 0.05). Conclusively, H3K27ac regulates genes involved in UL onset and maintenance. Histone deacetylation reversion upregulates the expression of tumor suppressor genes in UL cells, suggesting targeting histone modifications as a therapeutic approach for UL.
- Published
- 2022
14. Overall survival results from ICON8, a GCIG phase 3 randomised controlled trial assessing weekly dose-dense chemotherapy in first-line epithelial ovarian, fallopian tube, or primary peritoneal cancer treatment
- Author
-
McNeish, I, Clamp, A, James, EC, Dean, A, Kim, J-W, O’Donnell, DM, Gallardo-Rincon, D, Blagden, S, Brenton, J, Perren, TJ, Sundar, S, Lord, R, Dark, G, Hall, M, Banerjee, S, Glasspool, RM, Hanna, L, Williams, S, Scratchard, KM, Nam, H, Essapen, S, Parkinson, C, McAvan, L, Swart, AM, Popoola, B, Schiavone, F, Badrock, J, Fananapazir, F, Cook, AD, Parmar, M, Kaplan, R, Ledermann, JA, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Background: Standard of care first line chemotherapy for epithelial ovarian cancer (EOC) is carboplatin and paclitaxel administered once every three weeks. The JGOG3016 trial reported significant improvement in progression-free (PFS) and overall survival (OS) with dose-dense weekly paclitaxel and three-weekly carboplatin. However, this benefit was not observed in the progression free survival results of ICON8, previously reported. Here, we present the final co-primary OS and updated PFS outcomes. Methods: Women aged 18 or above with newly diagnosed International Federation of Gynecology and Obstetrics (FIGO) stage IC-IV EOC were recruited. Patients were randomly assigned to one of three groups; group 1: 3-weekly carboplatin area under the curve (AUC) 5/6 and 3-weekly paclitaxel 175mg/m2, group 2: 3-weekly carboplatin AUC5/6 and weekly paclitaxel 80mg/m2, group 3: weekly carboplatin AUC2 and weekly paclitaxel 80mg/m2, all administered by intravenous infusion. Patients entered the trial after immediate primary surgery (IPS) or received delayed primary surgery (DPS) during chemotherapy. Randomisation was performed using minimisation with stratification factors of GCIG group, disease stage and outcome and timing of surgery. Co-primary outcomes were PFS and OS, with comparisons performed between group 2 and group 1, and group 3 and group 1. Intention-to-treat analyses were powered to detect a hazard ratio of 0.75 in favour of the experimental groups. The trial is registered on clinicaltrials.gov as NCT01654146 and controlled-trials.com asISRCTN10356387. Findings: Between 6 Jun 2011 and 28 Nov 2014, 1566 women were recruited; group 1 (N=522), group 2 (N=523) and group 3 (N=521). Baseline characteristics include median age 62, 69% high grade serous carcinoma, 72% stage IIIC-IV disease, 48% IPS. By March 2020, with a median follow up of 69 months (IQR 61-75 months), 324, 309 and 313 deaths had occurred in groups 1, 2 and 3 respectively. No significant difference in OS was observed in either comparison: HR 0.87 (97.5% CI 0.73, 1.05) group 2 vs group 1, HR 0.91 (97.5% CI 0.76, 1.09) group 3 vs group 1. Most common G3/4 adverse events were reduced neutrophils (78, 183, 154 in groups 1, 2, 3) reduced white blood cell count (22, 80, 71 In groups 1, 2, 3) and anaemia (26, 66, 25 in groups 1, 2, 3). No new serious adverse events were reported. Interpretation: First-line weekly dose-dense chemotherapy does not improve OS for patients with EOC compared with standard 3-weekly chemotherapy and should not be used as part of standard multimodality ovarian cancer treatment for Caucasian women in the front-line setting. Funding: Cancer Research UK, Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, Cancer Australia.
- Published
- 2022
15. Cancer therapy approval timings, review speed and publication of pivotal registration trials in the US and Europe from 2010-2019
- Author
-
Mark P. Lythgoe, Aakash Desai, Bishal Gyawali, Philip Savage, Jonathan Krell, Jeremy L. Warner, Ali Raza Khaki, 4D Pharma Plc, and Ovarian Cancer Action
- Subjects
Europe ,Cross-Sectional Studies ,United States Food and Drug Administration ,Neoplasms ,Humans ,General Medicine ,Drug Approval ,United States - Abstract
Ensuring patients have access to safe and efficacious medicines in a timely manner is an essential goal for regulatory agencies, one which has particular importance in oncology because of the substantial unmet need for new therapies. The 2 largest regulatory agencies, the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), have pivotal global roles, and their recommendations and approvals are frequently followed by other national regulators.To compare market authorization dates for new oncology therapies approved in the US and Europe over the past decade and to examine and contrast the regulatory activities of the FDA and EMA in the approval of new cancer medicines.This cross-sectional study reviewed the FDA and EMA regulatory databases to identify new oncology therapies approved in both the US and Europe from 2010 to 2019, and characterization of the timings of regulatory activities. Statistical analysis was performed from January to April 2022.Regulatory approval date, review time, submission of market authorization application, accelerated approval or conditional marketing authorization status and proportion of approvals prior to peer-reviewed publication of pivotal trial results.In total, 89 new concomitant oncology therapies were approved in the US and Europe from 2010 to 2019. The FDA approved 85 oncology therapies (95%) before European authorization and 4 therapies (5%) after. The median (IQR) delay in market authorization for new oncology therapies in Europe was 241 (150-370) days compared with the US. The median (IQR) review time was 200 (155-277) days for the FDA and 426 (358-480) days for the EMA. Sixty-four new licensing applications (72%) were submitted to the FDA first, compared with 21 (23%) to the EMA. Thirty-five oncology therapies (39%) were approved by the FDA prior to pivotal study publication, whereas only 8 (9%) by the EMA.In this cross-sectional study, new oncology therapies were approved earlier in the US than Europe. The FDA received licensing applications sooner and had shorter review times. However, more therapies were approved prior to licensing study publication, leaving uncertainty for practitioners regarding clinical utility and safety of newly approved therapies.
- Published
- 2022
16. DICE: Dual mTorc Inhibition in advanCed/recurrent Epithelial ovarian cancer resistant to standard treatment—a study protocol for a randomised trial investigating a novel therapy called TAK228
- Author
-
Francesca Fiorentino, Jonathan Krell, Consuelo Nohpal de la Rosa, Lee Webber, Millennium Pharmaceuticals, Inc., and Ovarian Cancer Action
- Subjects
Ovarian Neoplasms ,Science & Technology ,Paclitaxel ,TAK228 ,Weekly paclitaxel ,Medicine (miscellaneous) ,1103 Clinical Sciences ,Research & Experimental Medicine ,Carcinoma, Ovarian Epithelial ,Medicine, Research & Experimental ,Cardiovascular System & Hematology ,Ovarian cancer ,General & Internal Medicine ,Antineoplastic Combined Chemotherapy Protocols ,PATTERNS ,Quality of Life ,Humans ,Female ,Pharmacology (medical) ,RECURRENT ,Platinum-resistant ,Neoplasm Recurrence, Local ,Life Sciences & Biomedicine ,1102 Cardiorespiratory Medicine and Haematology - Abstract
Background The standard initial treatment for ovarian cancer is surgery and platinum-based chemotherapy and potentially maintenance therapy with avastin or inhibitors of poly-ADP ribose polymerase (PARP). While a proportion of women are cured by this approach, the vast majority will relapse and become resistant to platinum chemotherapy either initially or on subsequent treatment. There is an unmet need to improve response to treatment and quality of life in these women. TAK228 is a novel therapy that can be added to standard treatment in the participant population and the aim of the DICE trial is to assess its effectiveness. Laboratory and clinical research has shown that these ovarian cancers may respond to the molecular target of a drug such as TAK228, and there have been studies using it in other advanced solid tumours including endometrial cancer. Methods One hundred twenty-four eligible women will be recruited from participating research sites in the United Kingdom (UK) and Germany. Randomised participants will receive either weekly paclitaxel alone (standard treatment, n = 62) or TAK228 plus weekly paclitaxel (n = 62) until the cancer significantly worsens; there are significant adverse events or any other protocol-defined stopping criteria. Participants will be monitored for response to treatment (using radiological imaging), adverse events and quality of life during both randomised treatment and subsequent follow-up. Discussion The primary objective/endpoint of the study is to compare the two treatments in terms of progression-free survival, or the length of time that each participant is alive without the cancer significantly worsening according to defined assessment criteria. If the addition of TAK228 to weekly paclitaxel chemotherapy is shown to significantly improve this statistically, and adverse events and quality of life are not significantly worse than standard treatment, then TAK228 plus weekly paclitaxel could potentially be taken forward within the context of a larger phase III trial. Trial registration ClinicalTrials.govNCT03648489. Registered on 27 August 2018.
- Published
- 2022
17. Development and Validation of the Gene Expression Predictor of High-grade Serous Ovarian Carcinoma Molecular SubTYPE (PrOTYPE)
- Author
-
Sven Mahner, Robertson Mackenzie, Aline Talhouk, Linda E. Kelemen, Gottfried E. Konecny, Jennifer Alsop, Rosalind Glasspool, Chiu-Chen Tseng, Joy Hendley, Dennis J. Slamon, Jennifer A. Doherty, Andrew Berchuck, Anna H. Wu, Anna M. Piskorz, Chen Wang, Cristina Rodríguez-Antona, D.G.H. de Silva, Valerie Rhenius, Peter A. Fasching, Stacey J. Winham, Gary L. Keeney, Teodora Goranova, Joshy George, Jan Lubinski, Michelle J. Henderson, Rex C. Bentley, Jenny Lester, Sabine Behrens, Joellen M. Schildkraut, Michael E. Carney, Timothy Budden, David G. Huntsman, Oleg Oszurek, Michael S. Anglesio, Jacek Gronwald, Ruby Yun-Ju Huang, Martin Köbel, Javier Benitez, Martin Widschwendter, Melissa C. Larson, Raghwa Sharma, Clara Bodelon, Usha Menon, Janusz Menkiszak, Blake Gilks, María Josefa Mosteiro García, Jesús García-Donas, Wafaa Elatre, Scott H. Kaufmann, Paul Haluska, Pamela J. Thompson, Boris Winterhoff, Susan J. Ramus, Louise A. Brinton, Simon A. Gayther, Mary Anne Rossing, Georgia Chenevix-Trench, Hugh Luk, Jolanta Lissowska, Marc T. Goodman, Billy Chen, Beth Y. Karlan, Naveena Singh, Sian Fereday, Mark E. Sherman, Ana Osorio, Lynne R. Wilkens, Maria P. Intermaggio, Brenda Y. Hernandez, Britton Trabert, Esther Herpel, Mercedes Jimenez-Linan, Janine Senz, Geyi Liu, Celeste Leigh Pearce, Samuel C Y Leong, Iain A. McNeish, Isabelle Ray-Coquard, Susana Banerjee, Malcolm C. Pike, Liz-Anne Lewsley, Helen Steed, Honglin Song, Samantha Hinsley, David D.L. Bowtell, James D. Brenton, Holly R. Harris, Tuan Zea Tan, Cezary Cybulski, Alicia Beeghly-Fadiel, A. Toloczko, Nikilyn Nevins, Robert S. Brown, Darren Ennis, Stephanie Chen, Euan A. Stronach, José Palacios, Sandra Orsulic, Anna deFazio, Geoff Macintyre, Kara L. Cushing-Haugen, Mila Volchek, Aleksandra Gentry-Maharaj, Jenny Chang-Claude, Ellen L. Goode, Paul D.P. Pharoah, Hanwei Sudderuddin, Stefan Kommoss, Derek S. Chiu, Huei San Leong, Peter Sinn, Catherine J. Kennedy, Chloe Karpinskyj, Alison Brand, Amy Lum, Veronica Chow, Nicolas Wentzensen, Tayyebeh M. Nazeran, Nadia Traficante, Dustin Johnson, Yoke-Eng Chiew, Casey S. Greene, Jennifer M Koziak, Renée T. Fortner, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, Ovarian Cancer Action, Talhouk, Aline [0000-0001-7760-410X], George, Joshy [0000-0001-8510-8229], Wang, Chen [0000-0003-2638-3081], Tan, Tuan Zea [0000-0001-6624-1593], Behrens, Sabine [0000-0002-9714-104X], Bodelon, Clara [0000-0002-6578-2678], Brinton, Louise [0000-0003-3853-8562], Fortner, Renée T [0000-0002-1426-8505], García-Donas, Jesús [0000-0001-7731-3601], Gentry-Maharaj, Aleksandra [0000-0001-7270-9762], Glasspool, Rosalind [0000-0002-5000-1680], Greene, Casey S [0000-0001-8713-9213], Harris, Holly R [0000-0002-2572-6727], Kaufmann, Scott H [0000-0002-4900-7145], Kennedy, Catherine J [0000-0002-4465-5784], Köbel, Martin [0000-0002-6615-2037], Koziak, Jennifer M [0000-0001-5830-0397], Lissowska, Jolanta [0000-0003-2695-5799], McNeish, Iain A [0000-0002-9387-7586], Menkiszak, Janusz [0000-0001-8279-7196], Hinsley, Samantha [0000-0001-6903-4688], Pike, Malcolm C [0000-0003-4891-1199], Rodriguez-Antona, Cristina [0000-0001-8750-7338], Sinn, Peter [0000-0003-2836-6699], Trabert, Britton [0000-0002-1539-6090], Widschwendter, Martin [0000-0002-7778-8380], Winham, Stacey J [0000-0002-8492-9102], Brenton, James D [0000-0002-5738-6683], Brown, Robert [0000-0001-7960-5755], Chang-Claude, Jenny [0000-0001-8919-1971], deFazio, Anna [0000-0003-0057-4744], Fasching, Peter A [0000-0003-4885-8471], Kelemen, Linda E [0000-0003-4362-9784], Menon, Usha [0000-0003-3708-1732], Pharoah, Paul DP [0000-0001-8494-732X], Ramus, Susan J [0000-0003-0005-7798], Doherty, Jennifer A [0000-0002-1454-8187], Anglesio, Michael S [0000-0003-1639-5003], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Bevacizumab ,03 medical and health sciences ,Ovarian tumor ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Ovarian carcinoma ,Internal medicine ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Stage (cooking) ,Aged ,Ovarian Neoplasms ,business.industry ,Cystadenoma, Serous ,Cancer ,Middle Aged ,Precision medicine ,Omics ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Serous fluid ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,Transcriptome ,business ,Algorithms ,medicine.drug - Abstract
Purpose: Gene expression–based molecular subtypes of high-grade serous tubo-ovarian cancer (HGSOC), demonstrated across multiple studies, may provide improved stratification for molecularly targeted trials. However, evaluation of clinical utility has been hindered by nonstandardized methods, which are not applicable in a clinical setting. We sought to generate a clinical grade minimal gene set assay for classification of individual tumor specimens into HGSOC subtypes and confirm previously reported subtype-associated features. Experimental Design: Adopting two independent approaches, we derived and internally validated algorithms for subtype prediction using published gene expression data from 1,650 tumors. We applied resulting models to NanoString data on 3,829 HGSOCs from the Ovarian Tumor Tissue Analysis consortium. We further developed, confirmed, and validated a reduced, minimal gene set predictor, with methods suitable for a single-patient setting. Results: Gene expression data were used to derive the predictor of high-grade serous ovarian carcinoma molecular subtype (PrOTYPE) assay. We established a de facto standard as a consensus of two parallel approaches. PrOTYPE subtypes are significantly associated with age, stage, residual disease, tumor-infiltrating lymphocytes, and outcome. The locked-down clinical grade PrOTYPE test includes a model with 55 genes that predicted gene expression subtype with >95% accuracy that was maintained in all analytic and biological validations. Conclusions: We validated the PrOTYPE assay following the Institute of Medicine guidelines for the development of omics-based tests. This fully defined and locked-down clinical grade assay will enable trial design with molecular subtype stratification and allow for objective assessment of the predictive value of HGSOC molecular subtypes in precision medicine applications. See related commentary by McMullen et al., p. 5271
- Published
- 2020
18. Weekly platinum-based chemotherapy versus 3-weekly platinum-based chemotherapy for newly diagnosed ovarian cancer (ICON8): quality-of-life results of a phase 3, randomised, controlled trial
- Author
-
Timothy J. Perren, Rosemary Lord, Jane Hook, Ian A. McNeish, Christopher J. Poole, Jonathan A. Ledermann, Helena M. Earl, Sarah P. Blagden, Adrian Cook, Jae Weon Kim, Graham Dark, Andrew R Clamp, Marcia Hall, Dearbhaile M. O'Donnell, Richard Kaplan, Ian R. White, Lesley Howells, Andrew Dean, Elizabeth C. James, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and Cancer Research UK
- Subjects
Cross-sectional study ,medicine.medical_treatment ,EUROPEAN-ORGANIZATION ,Carcinoma, Ovarian Epithelial ,Carboplatin ,law.invention ,chemistry.chemical_compound ,0302 clinical medicine ,Randomized controlled trial ,law ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,030212 general & internal medicine ,Young adult ,Aged, 80 and over ,Ovarian Neoplasms ,QLQ-C30 ,Middle Aged ,Prognosis ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,Female ,Life Sciences & Biomedicine ,Adult ,medicine.medical_specialty ,Adolescent ,Paclitaxel ,QUESTIONNAIRE ,Drug Administration Schedule ,Article ,Young Adult ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Survival rate ,Platinum ,Aged ,Chemotherapy ,Science & Technology ,business.industry ,Clinical trial ,Cross-Sectional Studies ,chemistry ,Quality of Life ,business ,Follow-Up Studies - Abstract
Summary Background The ICON8 study reported no significant improvement in progression-free survival (a primary endpoint) with weekly chemotherapy compared with standard 3-weekly treatment among patients with epithelial ovarian cancer. All ICON8 patients were eligible to take part in the accompanying health-related quality-of-life study, which measured the effect of treatment on self-reported wellbeing, reported here. Methods In this open-label, randomised, controlled, phase 3, three-arm, Gynecologic Cancer Intergroup (GCIG) trial done at 117 hospital sites in the UK, Australia, New Zealand, Mexico, South Korea, and Republic of Ireland, women (aged at least 18 years) with newly diagnosed, histologically confirmed International Federation of Gynecology and Obstetrics stage IC–IV ovarian cancer and an Eastern Cooperative Oncology Group performance status of 0–2 were randomly assigned (1:1:1) centrally using minimisation to group 1 (intravenous carboplatin area under the curve [AUC]5 or AUC6 and 175 mg/m2 intravenous paclitaxel every 3 weeks), group 2 (carboplatin AUC5 or AUC6 every 3 weeks and 80 mg/m2 paclitaxel weekly), or group 3 (carboplatin AUC2 weekly and 80 mg/m2 paclitaxel weekly). Randomisation was stratified by GCIG group, disease stage, and outcome and timing of surgery. Patients and clinicians were not masked to treatment assignment. Patients underwent immediate or delayed primary surgery according to clinicians' choice. Patients were asked to complete European Organisation for Research and Treatment of Cancer QLQ-C30 and QLQ-OV28 questionnaires at enrolment, before each chemotherapy cycle, then 6-weekly up to 9 months, 3-monthly up to 2 years, and 6-monthly up to 5 years. Quality of life was a prespecified secondary outcome of the ICON8 study. Within the quality-of-life study, the co-primary endpoints were QLQ-C30 global health score at 9 months (cross-sectional analysis) and mean QLQ-C30 global health score from randomisation to 9 months (longitudinal analysis). Data analyses were done on an intention-to-treat basis. The trial is registered on ClinicalTrials.gov , NCT01654146 and ISRCTN Registry, ISRCTN10356387, and is currently in long-term follow up. Findings Between June 6, 2011, and Nov 28, 2014, 1566 patients were recruited into ICON8 (522 were included in group 1, 523 in group 2, and 521 in group 3). Baseline quality-of-life questionnaires were completed by 1438 (92%) of 1566 patients and 9-month questionnaires by 882 (69%) of 1280 patients. We observed no significant difference in global health score at 9 months (cross-sectional analysis) between study groups (group 2 vs group 1, difference in mean score 2·3, 95% CI −0·4 to 4·9, p=0·095; group 3 vs group 1, −0·8, −3·8 to 2·2, p=0·61). Using longitudinal analysis, we found lower global health scores for those receiving weekly paclitaxel than for those receiving 3-weekly chemotherapy (group 2 vs group 1, mean difference −1·8, 95% CI −3·6 to −0·1, p=0·043; group 3 vs group 1, −2·9, −4·7 to −1·1, p=0·0018). Interpretation We found no evidence of a difference in global quality of life between treatment groups at 9 months; however, patients receiving weekly treatment reported lower mean quality of life across the 9-month period after randomisation. Taken together with the lack of progression-free survival benefit, these findings do not support routine use of weekly paclitaxel-containing regimens in the management of newly diagnosed ovarian cancer. Funding Cancer Research UK, Medical Research Council, Health Research Board Ireland, Irish Cancer Society, and Cancer Australia.
- Published
- 2020
19. Proteomic analysis of malignant and benign endometrium according to obesity and insulin-resistance status using Reverse Phase Protein Array
- Author
-
Maria Kyrgiou, Marc J. Gunter, Haonan Lu, Jaya Nautiyal, Olivia Raglan, Nada Assi, Hani Gabra, Ovarian Cancer Action, and HCA International Limited
- Subjects
Adult ,Proteomics ,0301 basic medicine ,medicine.medical_treatment ,Protein Array Analysis ,Endometrium ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Downregulation and upregulation ,Physiology (medical) ,Hyperinsulinemia ,medicine ,Humans ,Obesity ,General Clinical Medicine ,Aged ,Aged, 80 and over ,business.industry ,Insulin ,Endometrial cancer ,Biochemistry (medical) ,Public Health, Environmental and Occupational Health ,Reverse phase protein lysate microarray ,1103 Clinical Sciences ,General Medicine ,Middle Aged ,medicine.disease ,Endometrial Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Insulin Resistance ,business ,Signal Transduction - Abstract
Obesity and hyperinsulinemia are known risk factors for endometrial cancer, yet thebiological pathways underlying this relationship are incompletely understood. Thisstudy investigated protein expression in endometrial cancer and benign tissue andits correlation with obesity and insulin resistance.One hundred and seven women undergoing hysterectomy for endometrial canceror benign conditions provided a fasting blood sample and endometrial tissue. Weperformed proteomic expression according to body mass index, insulin resistance,and serum marker levels. We used linear regression and independentttest for statis-tical analysis. Proteomic data from 560 endometrial cancer cases from The CancerGenome Atlas (TCGA) databank were used to assess reproducibility of results.One hundred and twenty seven proteins were significantly differentially expressedbetween 66 cancer and 26 benign patients. Protein expression involved in cellcycle progression, impacting cytoskeletal dynamics (PAK1) and cell survival (Rab25), were most significantly altered. Obese women with cancer had increasedPRAS40_pT246; a downstream marker of increased PI3K-AKT signaling. Obesewomen without cancer had increased mitogenic and antiapoptotic signaling byway of upregulation of Mcl-1, DUSP4, and Insulin Receptor-b.This exploratory study identified a number of candidate proteins specific to endo-metrioid endometrial cancer and benign endometrial tissues. Obesity and insulinresistance in women with benign endometrium leads to specific upregulation ofproteins involved in insulin and driver oncogenic signaling pathways such as thePI3K-AKT-mTOR and growth factor signaling pathways which are mitogenic andalso disruptive to metabolism. (Translational Research 2020; 000:1 16)
- Published
- 2020
20. Cancer-Specific Loss of p53 Leads to a Modulation of Myeloid and T Cell Responses
- Author
-
Charles Swanton, Marc Hennequart, Sebastijan Hobor, Julianna Blagih, Ming Yang, Susan M. Mason, Steven Pilley, Josephine Walton, Karen H. Vousden, Fabio Zani, Karen Blyth, Jennifer P. Morton, Probir Chakravarty, Andreas K. Hock, Iain A. McNeish, Eva Grönroos, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
p53 ,Myeloid ,PROMOTES ,BLOCKADE ,PROGRESSION ,MICROENVIRONMENT ,0601 Biochemistry and Cell Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Immune tolerance ,Mice ,0302 clinical medicine ,Ecology,Evolution & Ethology ,Macrophage ,MACROPHAGES ,lcsh:QH301-705.5 ,Chemical Biology & High Throughput ,Human Biology & Physiology ,0303 health sciences ,Genome Integrity & Repair ,3. Good health ,medicine.anatomical_structure ,myeloid cells ,030220 oncology & carcinogenesis ,GROWTH ,Genetics & Genomics ,Life Sciences & Biomedicine ,tumor ,T cell ,IMMUNITY ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Signalling & Oncogenes ,03 medical and health sciences ,Immune system ,INFLAMMATION ,medicine ,Animals ,Humans ,Computational & Systems Biology ,030304 developmental biology ,Science & Technology ,Cell Biology ,Tumour Biology ,TUMOR-SUPPRESSOR P53 ,T cell response ,Metabolism ,lcsh:Biology (General) ,1116 Medical Physiology ,Cancer cell ,Kras ,Cancer research ,Tumor Suppressor Protein p53 ,Carcinogenesis ,CD8 ,RAS - Abstract
Summary Loss of p53 function contributes to the development of many cancers. While cell-autonomous consequences of p53 mutation have been studied extensively, the role of p53 in regulating the anti-tumor immune response is still poorly understood. Here, we show that loss of p53 in cancer cells modulates the tumor-immune landscape to circumvent immune destruction. Deletion of p53 promotes the recruitment and instruction of suppressive myeloid CD11b+ cells, in part through increased expression of CXCR3/CCR2-associated chemokines and macrophage colony-stimulating factor (M-CSF), and attenuates the CD4+ T helper 1 (Th1) and CD8+ T cell responses in vivo. p53-null tumors also show an accumulation of suppressive regulatory T (Treg) cells. Finally, we show that two key drivers of tumorigenesis, activation of KRAS and deletion of p53, cooperate to promote immune tolerance., Graphical Abstract, Highlights • Tumor-specific loss of p53 delays tumor rejection in immune-competent hosts • p53 loss increases myeloid infiltration through enhanced cytokine secretion • The increase in Treg cells in response to loss of p53 is independent of Kras mutation • Kras mutations coordinate with p53 loss to regulate myeloid recruitment, TP53 is one of the most frequently mutated genes in cancer; however, its significance in controlling tumor-immune crosstalk is not fully understood. Blagih et al. highlight a key role for tumor-associated loss of p53, a common oncogenic event, in regulating myeloid and T cell responses.
- Published
- 2020
21. Sixth ovarian cancer consensus conference on clinical research of the gynecologic cancer InterGroup
- Author
-
McNeish, I, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
The Gynecologic Cancer InterGroup (GCIG) sixth Ovarian Cancer Conference on Clinical Research (OCCC6) was held virtually in October 2021 following published consensus guidelines. All 33 GCIG member groups participated in the development, refinement, and adoption of 20 statements within 4 topic groups on clinical research including first line treatment, recurrent disease, disease subgroups and future trials. Unanimous consensus was obtained for 14 of 20 statements, with >90% concordance in the remaining 6 statements. The high acceptance rate following active deliberation amongst the GCIG groups confirmed that a consensus process could be applied in a virtual setting. Together with detailed categorisation of unmet needs, these consensus statements will promote harmonisation of international clinical research in ovarian cancer.
- Published
- 2022
22. Polygenic risk modeling for prediction of epithelial ovarian cancer risk
- Author
-
Dareng, Eileen O., Tyrer, Jonathan P., Barnes, Daniel R., Jones, Michelle R., Yang, Xin, Aben, Katja K. H., Adank, Muriel A., Agata, Simona, Andrulis, Irene L., Anton-Culver, Hoda, Antonenkova, Natalia N., Aravantinos, Gerasimos, Arun, Banu K., Augustinsson, Annelie, Balmaña, Judith, Bandera, Elisa V., Barkardottir, Rosa B., Barrowdale, Daniel, Beckmann, Matthias W., Beeghly-Fadiel, Alicia, Benitez, Javier, Bermisheva, Marina, Bernardini, Marcus Q., Bjorge, Line, Black, Amanda, Bogdanova, Natalia V., Bonanni, Bernardo, Borg, Ake, Brenton, James D., Budzilowska, Agnieszka, Butzow, Ralf, Buys, Saundra S., Cai, Hui, Caligo, Maria A., Campbell, Ian, Cannioto, Rikki, Cassingham, Hayley, Chang-Claude, Jenny, Chanock, Stephen J., Chen, Kexin, Chiew, Yoke-Eng, Chung, Wendy K., Claes, Kathleen, Colonna, Sarah, Lesueur, Fabienne, Mebirouk, Noura, Engel, Christoph, Schmutzler, Rita K., Davies, Eleanor, Eccles, Diana M., Evans, D. Gareth, Cook, Linda S., Couch, Fergus J., Daly, Mary B., Dao, Fanny, de la Hoya, Miguel, de Putter, Robin, Dennis, Joe, DePersia, Allison, Devilee, Peter, Diez, Orland, Ding, Yuan Chun, Doherty, Jennifer A., Domchek, Susan M., Dörk, Thilo, du Bois, Andreas, Dürst, Matthias, Eliassen, Heather A., Evans, Gareth D., Fasching, Peter A., Flanagan, James M., Fortner, Renée T., Machackova, Eva, Friedman, Eitan, Ganz, Patricia A., Garber, Judy, Gensini, Francesca, Giles, Graham G., Glendon, Gord, Godwin, Andrew K., Goodman, Marc T., Greene, Mark H., Gronwald, Jacek, Hahnen, Eric, Haiman, Christopher A., Håkansson, Niclas, Hamann, Ute, Hansen, Thomas V. O., Harris, Holly R., Hartman, Mikael, Heitz, Florian, Hildebrandt, Michelle A. T., Høgdall, Estrid, Høgdall, Claus K., Hopper, John L., Huang, Ruea-Yea, Huff, Chad, Hulick, Peter J., Huntsman, David G., Imyanitov, Evgeny N., Chenevix-Trench, Georgia, van der Hout, Annemieke H., Isaacs, Claudine, Jakubowska, Anna, James, Paul A., Janavicius, Ramunas, Jensen, Allan, Johannsson, Oskar Th., John, Esther M., Jones, Michael E., Kang, Daehee, Karlan, Beth Y., Karnezis, Anthony, Kelemen, Linda E., Khusnutdinova, Elza, Kiemeney, Lambertus A., Kim, Byoung-Gie, Kjaer, Susanne K., Komenaka, Ian, Kupryjanczyk, Jolanta, Kurian, Allison W., Kwong, Ava, Lambrechts, Diether, Larson, Melissa C., Lazaro, Conxi, Le, Nhu D., Leslie, Goska, Lester, Jenny, Levine, Douglas A., Li, Lian, Li, Jingmei, Loud, Jennifer T., Lu, Karen H., Lubiński, Jan, Mai, Phuong L., Manoukian, Siranoush, Marks, Jeffrey R., Matsuno, Rayna Kim, Matsuo, Keitaro, May, Taymaa, McGuffog, Lesley, McLaughlin, John R., McNeish, Iain A., Menon, Usha, Miller, Austin, Milne, Roger L., Minlikeeva, Albina, Modugno, Francesmary, Montagna, Marco, Moysich, Kirsten B., Munro, Elizabeth, Nathanson, Katherine L., Neuhausen, Susan L., Nevanlinna, Heli, Yie, Joanne Ngeow Yuen, Nielsen, Henriette Roed, Nielsen, Finn C., Nikitina-Zake, Liene, Odunsi, Kunle, Offit, Kenneth, Olah, Edith, Olbrecht, Siel, Olopade, Olufunmilayo I., Olson, Sara H., Olsson, Håkan, Osorio, Ana, Papi, Laura, Park, Sue K., Parsons, Michael T., Pathak, Harsha, Pedersen, Inge Sokilde, Peixoto, Ana, Pejovic, Tanja, Perez-Segura, Pedro, Permuth, Jennifer B., Peshkin, Beth, Peterlongo, Paolo, Piskorz, Anna, Prokofyeva, Darya, Radice, Paolo, Rantala, Johanna, Riggan, Marjorie J., Risch, Harvey A., Rodriguez-Antona, Cristina, Ross, Eric, Rossing, Mary Anne, Runnebaum, Ingo, Sandler, Dale P., Santamariña, Marta, Soucy, Penny, Setiawan, V. Wendy, Shan, Kang, Sieh, Weiva, Simard, Jacques, Singer, Christian F., Sokolenko, Anna P., Song, Honglin, Southey, Melissa C., Steed, Helen, Stoppa-Lyonnet, Dominique, Sutphen, Rebecca, Swerdlow, Anthony J., Tan, Yen Yen, Teixeira, Manuel R., Teo, Soo Hwang, Terry, Kathryn L., Terry, Mary Beth, Thompson, Pamela J., Thomsen, Liv Cecilie Vestrheim, Titus, Linda, Trabert, Britton, Travis, Ruth, Tworoger, Shelley S., Valen, Ellen, van Altena, Anne M., Van Nieuwenhuysen, Els, Edwards, Digna Velez, Vierkant, Robert A., Wang, Frances, Webb, Penelope M., Weinberg, Clarice R., Wentzensen, Nicolas, White, Emily, Whittemore, Alice S., Winham, Stacey J., Wolk, Alicja, Woo, Yin-Ling, Wu, Anna H., Yan, Li, Yannoukakos, Drakoulis, Zheng, Wei, Ziogas, Argyrios, Lawrenson, Kate, deFazio, Anna, Ramus, Susan J., Pearce, Celeste L., Monteiro, Alvaro N., Cunningham, Julie M., Goode, Ellen L., Schildkraut, Joellen M., Berchuck, Andrew, Gayther, Simon A., Pharoah, Paul D. P., Claes, Kathleen B. M., Thomassen, Mads, Thull, Darcy L., Tischkowitz, Marc, Toland, Amanda E., Torres, Diana, Tung, Nadine, van Rensburg, Elizabeth J., Vega, Ana, Wappenschmidt, Barbara, Weitzel, Jeffrey N., Zavaglia, Katia M., Zorn, Kristin K., Sellers, Thomas A., Antoniou, Antonis C., Kleibl, Zdenek, Easton, Douglas, DeFazio, Anna, Cunningham, Julie, GEMO Study Collaborators, [missing], GC-HBOC Study Collaborators, [missing], EMBRACE Collaborators, [missing], OPAL Study Group, [missing], AOCS Group, [missing], KConFab Investigators, [missing], HEBON Investigators, [missing], The OCAC Consortium, [missing], The CIMBA Consortium, [missing], Clinicum, Department of Pathology, HUSLAB, Department of Obstetrics and Gynecology, HUS Gynecology and Obstetrics, Faculteit Medische Wetenschappen/UMCG, Institut Català de la Salut, [Dareng EO, Barnes DR, Yang X] University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK. [Tyrer JP] University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK. [Jones MR] Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA. [Aben KKH] Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands. Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands. [Balmaña J] Hereditary cancer Genetics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Diez O] Oncogenetics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Àrea de Genètica Clínica i Molecular, Vall d’Hebron Hospital Universitari, Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Male ,Biochemistry & Molecular Biology ,OCAC Consortium ,Genetic Phenomena::Genotype::Genetic Predisposition to Disease [PHENOMENA AND PROCESSES] ,Breast Neoplasms ,Ovaris - Càncer - Aspectes genètics ,Carcinoma, Ovarian Epithelial ,SUSCEPTIBILITY ,Polymorphism, Single Nucleotide ,GEMO Study Collaborators ,Risk Factors ,OPAL Study Group ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,Medicine and Health Sciences ,Genetics ,Ovaris - Càncer - Propensió ,Humans ,Genetic Predisposition to Disease ,Prospective Studies ,EMBRACE Collaborators ,GC-HBOC Study Collaborators ,Genetics (clinical) ,HEBON Investigators ,fenómenos genéticos::genotipo::predisposición genética a la enfermedad [FENÓMENOS Y PROCESOS] ,Medicinsk genetik ,Ovarian Neoplasms ,Genetics & Heredity ,0604 Genetics ,Science & Technology ,Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,1184 Genetics, developmental biology, physiology ,Neoplasms::Neoplasms::Neoplasms by Site::Endocrine Gland Neoplasms::Ovarian Neoplasms::Carcinoma, Ovarian Epithelial [DISEASES] ,1103 Clinical Sciences ,Bayes Theorem ,AOCS Group ,neoplasias::neoplasias::neoplasias por localización::neoplasias de las glándulas endocrinas::neoplasias ováricas::carcinoma epitelial de ovario [ENFERMEDADES] ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Carcinoma, Ovarian Epithelial/genetics ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,CIMBA Consortium ,1182 Biochemistry, cell and molecular biology ,Female ,KConFab Investigators ,Ovarian Neoplasms/epidemiology ,Medical Genetics ,Life Sciences & Biomedicine - Abstract
Clinical genetics; Genetic markers; Risk factors Genética clínica; Marcadores genéticos; Factores de riesgo Genètica clínica; Marcadors genètics; Factors de risc Polygenic risk scores (PRS) for epithelial ovarian cancer (EOC) have the potential to improve risk stratification. Joint estimation of Single Nucleotide Polymorphism (SNP) effects in models could improve predictive performance over standard approaches of PRS construction. Here, we implemented computationally efficient, penalized, logistic regression models (lasso, elastic net, stepwise) to individual level genotype data and a Bayesian framework with continuous shrinkage, “select and shrink for summary statistics” (S4), to summary level data for epithelial non-mucinous ovarian cancer risk prediction. We developed the models in a dataset consisting of 23,564 non-mucinous EOC cases and 40,138 controls participating in the Ovarian Cancer Association Consortium (OCAC) and validated the best models in three populations of different ancestries: prospective data from 198,101 women of European ancestries; 7,669 women of East Asian ancestries; 1,072 women of African ancestries, and in 18,915 BRCA1 and 12,337 BRCA2 pathogenic variant carriers of European ancestries. In the external validation data, the model with the strongest association for non-mucinous EOC risk derived from the OCAC model development data was the S4 model (27,240 SNPs) with odds ratios (OR) of 1.38 (95% CI: 1.28–1.48, AUC: 0.588) per unit standard deviation, in women of European ancestries; 1.14 (95% CI: 1.08–1.19, AUC: 0.538) in women of East Asian ancestries; 1.38 (95% CI: 1.21–1.58, AUC: 0.593) in women of African ancestries; hazard ratios of 1.36 (95% CI: 1.29–1.43, AUC: 0.592) in BRCA1 pathogenic variant carriers and 1.49 (95% CI: 1.35–1.64, AUC: 0.624) in BRCA2 pathogenic variant carriers. Incorporation of the S4 PRS in risk prediction models for ovarian cancer may have clinical utility in ovarian cancer prevention programs.
- Published
- 2022
23. Weekly dose-dense chemotherapy in first-line epithelial ovarian, fallopian tube, or primary peritoneal carcinoma treatment (ICON8): primary progression free survival analysis results from a GCIG phase 3 randomised controlled trial
- Author
-
Hani Gabra, Dearbhaile M. O'Donnell, S. P. Stenning, Christopher Coyle, Jane Hook, Rosalind Glasspool, Graham Dark, Rachel Jones, Susana Banerjee, Jonathan A. Ledermann, Helena M. Earl, Andrew R Clamp, Adrian Cook, Marcia Hall, Timothy J. Perren, Sarah Williams, Rosemary Lord, Mahesh K. B. Parmar, Gosala S. Gopalakrishnan, Ann Marie Swart, Jae Weon Kim, Sudha Sundar, James D. Brenton, Elizabeth C. James, Raj Naik, Richard Kaplan, Iain A. McNeish, Andrew Dean, Sarah P. Blagden, Imperial College Healthcare NHS Trust- BRC Funding, and Ovarian Cancer Action
- Subjects
medicine.medical_treatment ,Carcinoma, Ovarian Epithelial ,030204 cardiovascular system & hematology ,Carboplatin ,law.invention ,chemistry.chemical_compound ,Gynecologic Surgical Procedures ,0302 clinical medicine ,Primary peritoneal carcinoma ,Randomized controlled trial ,law ,Antineoplastic Combined Chemotherapy Protocols ,030212 general & internal medicine ,Chemotherapy-Induced Febrile Neutropenia ,Peritoneal Neoplasms ,11 Medical and Health Sciences ,Ovarian Neoplasms ,Peripheral Nervous System Diseases ,Cytoreduction Surgical Procedures ,General Medicine ,Middle Aged ,Neoadjuvant Therapy ,Progression-Free Survival ,Chemotherapy, Adjuvant ,Female ,medicine.medical_specialty ,Paclitaxel ,Antineoplastic Agents ,White People ,Article ,03 medical and health sciences ,Asian People ,General & Internal Medicine ,Internal medicine ,medicine ,Fallopian Tube Neoplasms ,Animals ,Humans ,Progression-free survival ,Fallopian Tubes ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Chemotherapy ,business.industry ,Carcinoma ,medicine.disease ,Clinical trial ,Regimen ,chemistry ,Neoplasm Grading ,business ,Febrile neutropenia - Abstract
Background:\ud Carboplatin and paclitaxel administered every 3 weeks is standard-of-care first-line chemotherapy for epithelial ovarian cancer. The Japanese JGOG3016 trial showed a significant improvement in progression-free and overall survival with dose-dense weekly paclitaxel and 3-weekly carboplatin. In this study, we aimed to compare efficacy and safety of two dose-dense weekly regimens to standard 3-weekly chemotherapy in a predominantly European population with epithelial ovarian cancer.\ud Methods:\ud In this phase 3 trial, women with newly diagnosed International Federation of Gynecology and Obstetrics stage IC–IV epithelial ovarian cancer were randomly assigned to group 1 (carboplatin area under the curve [AUC]5 or AUC6 and 175 mg/m2 paclitaxel every 3 weeks), group 2 (carboplatin AUC5 or AUC6 every 3 weeks and 80 mg/m2 paclitaxel weekly), or group 3 (carboplatin AUC2 and 80 mg/m2 paclitaxel weekly). Written informed consent was provided by all women who entered the trial. The protocol had the appropriate national research ethics committee approval for the countries where the study was conducted. Patients entered the trial after immediate primary surgery, or before neoadjuvant chemotherapy with subsequent planned delayed primary surgery. The trial coprimary outcomes were progression-free survival and overall survival. Data analyses were done on an intention-to-treat basis, and were powered to detect a hazard ratio of 0·75 in progression-free survival. The main comparisons were between the control group (group 1) and each of the weekly research groups (groups 2 and 3).\ud Findings:\ud Between June 6, 2011, and Nov 28, 2014, 1566 women were randomly assigned to treatment. 72% (365), completed six protocol-defined treatment cycles in group 1, 60% (305) in group 2, and 63% (322) in group 3, although 90% (454), 89% (454), and 85% (437) completed six platinum-based chemotherapy cycles, respectively. Paclitaxel dose intensification was achieved with weekly treatment (median total paclitaxel dose 1010 mg/m2 in group 1; 1233 mg/m2 in group 2; 1274 mg/m2 in group 3). By February, 2017, 1018 (65%) patients had experienced disease progression. No significant progression-free survival increase was observed with either weekly regimen (restricted mean survival time 24·4 months [97·5% CI 23·0–26·0] in group 1, 24·9 months [24·0–25·9] in group 2, 25·3 months [23·9–26·9] in group 3; median progression-free survival 17·7 months [IQR 10·6–not reached] in group 1, 20·8 months [11·9–59·0] in group 2, 21·0 months [12·0–54·0] in group 3; log-rank p=0·35 for group 2 vs group 1; group 3 vs 1 p=0·51). Although grade 3 or 4 toxic effects increased with weekly treatment, these effects were predominantly uncomplicated. Febrile neutropenia and sensory neuropathy incidences were similar across groups.\ud Interpretation:\ud Weekly dose-dense chemotherapy can be delivered successfully as first-line treatment for epithelial ovarian cancer but does not significantly improve progression-free survival compared with standard 3-weekly chemotherapy in predominantly European populations.\ud Funding:\ud Cancer Research UK, Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, Cancer Australia.
- Published
- 2019
24. The association between obesity and weight loss after bariatric surgery on the vaginal microbiota
- Author
-
Hani Gabra, Anita Mitra, Yun S. Lee, Maria Kyrgiou, Olivia Raglan, David A. MacIntyre, Sanjay Purkayastha, Nada Assi, Phillip R. Bennett, Ann Smith, Jaya Nautiyal, Marc J. Gunter, Julian Marchesi, Imperial Health Charity, Ovarian Cancer Action, HCA International Limited, and Imperial College Healthcare Charity
- Subjects
MENOPAUSE STATUS ,Prevotella ,Overweight ,Microbial ecology ,0302 clinical medicine ,BACTERIAL-VAGINOSIS ,Weight loss ,1108 Medical Microbiology ,Lactobacillus ,RNA, Ribosomal, 16S ,Body mass index ,0303 health sciences ,Lactobacillus crispatus ,PRETERM ,Microbiota ,QR100-130 ,POLYCYSTIC-OVARY-SYNDROME ,PREGNANCY ,030220 oncology & carcinogenesis ,Vagina ,Female ,Vaginal microbiota ,Bacterial vaginosis ,medicine.symptom ,Life Sciences & Biomedicine ,0605 Microbiology ,Microbiology (medical) ,medicine.medical_specialty ,CANCER-RISK ,Firmicutes ,Biology ,Microbiology ,03 medical and health sciences ,BMI ,INFLAMMATION ,CONCEPTION ,Weight Loss ,medicine ,Humans ,Obesity ,030304 developmental biology ,Bariatric surgery ,Science & Technology ,0602 Ecology ,Research ,CERVICAL INTRAEPITHELIAL NEOPLASIA ,biology.organism_classification ,medicine.disease ,Surgery ,BODY-MASS INDEX ,Dysbiosis - Abstract
BackgroundObesity and vaginal microbiome (VMB) dysbiosis are each risk factors for adverse reproductive and oncological health outcomes in women. Here, we investigated the relationship between obesity, vaginal bacterial composition, local inflammation and bariatric surgery.MethodsVaginal bacterial composition assessed by high-throughput sequencing of bacterial 16S rRNA genes and local cytokine levels measured using a multiplexed Magnetic Luminex Screening Assay were compared between 67 obese and 42 non-obese women. We further assessed temporal changes in the microbiota and cytokines in a subset of 27 women who underwent bariatric surgery.ResultsThe bacterial component of the vaginal microbiota in obese women was characterised by a lower prevalence of aLactobacillus-dominant VMB and higher prevalence of a high diversity (Lactobacillusspp., andGardnerella- spp. depleted) VMB, compared with non-obese subjects (pDialisterspecies (pAnaerococcus vaginalis(p=0.021), andPrevotella timonensis(p=0.020) and decreased relative abundance ofLactobacillus crispatus(p=0.014). Local vaginal IL-1β, IL-4, IL-6, IL-8, IFNγ, MIP-1α and TNFα levels were all higher among obese women, however, only IL-1β and IL-8 correlated with VMB species diversity. In a subset of obese women undergoing bariatric surgery, there were no significant overall differences in VMB following surgery; however, 75% of these women remained obese at 6 months. Prior to surgery, there was no relationship between body mass index (BMI) and VMB structure; however, post-surgery women with aLactobacillus-dominant VMB had a significantly lower BMI than those with a high diversity VMB.ConclusionsObese women have a significantly different vaginal microbiota composition with increased levels of local inflammation compared to non-obese women. Bariatric surgery does not change the VMB; however, those with the greatest weight loss 6-month post-surgery are most likely to have aLactobacillus-dominant VMB.
- Published
- 2021
25. Surgery due to mechanical bowel obstruction in relapsed ovarian cancer: clinical and surgical results of a bicentric analysis of 87 patients (Oct, 10.1007/s00404-021-06237-x, 2021)
- Author
-
Armbrust, R, Chekerov, R, Sander, S, Biebl, M, Chopra, S, Krell, J, Rinne, N, Nixon, K, Fotopoulou, C, Sehouli, J, and Ovarian Cancer Action
- Subjects
Science & Technology ,Obstetrics & Gynecology ,1114 Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Life Sciences & Biomedicine - Published
- 2021
26. The Genomic Landscape of Early-Stage Ovarian High-Grade Serous Carcinoma
- Author
-
Cheng, Z, Mirza, H, Ennis, DP, Smith, P, Morrill Gavarro, L, Sokota, C, Giannone, G, Goranova, T, Bradley, T, Piskorz, A, Lockley, M, For the BriTROC-1 Investigators, Kaur, B, Singh, N, Tookman, L, Krell, J, McDermott, J, Macintyre, G, Markowetz, F, Brenton, JD, McNeish, I, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Ovarian Neoplasms ,Cancer Research ,BriTROC-1 Investigators ,High-Throughput Nucleotide Sequencing ,Genomics ,Carcinoma, Ovarian Epithelial ,Middle Aged ,Article ,Cystadenocarcinoma, Serous ,Oncology ,Mutation ,Fallopian Tube Neoplasms ,Humans ,1112 Oncology and Carcinogenesis ,Female ,Oncology & Carcinogenesis - Abstract
Purpose: Ovarian high-grade serous carcinoma (HGSC) is usually diagnosed at late stage. We investigated whether late-stage HGSC has unique genomic characteristics consistent with acquisition of evolutionary advantage compared with early-stage tumors. Experimental Design: We performed targeted next-generation sequencing and shallow whole-genome sequencing (sWGS) on pretreatment samples from 43 patients with FIGO stage I–IIA HGSC to investigate somatic mutations and copy-number (CN) alterations (SCNA). We compared results to pretreatment samples from 52 patients with stage IIIC/IV HGSC from the BriTROC-1 study. Results: Age of diagnosis did not differ between early-stage and late-stage patients (median 61.3 years vs. 62.3 years, respectively). TP53 mutations were near-universal in both cohorts (89% early-stage, 100% late-stage), and there were no significant differences in the rates of other somatic mutations, including BRCA1 and BRCA2. We also did not observe cohort-specific focal SCNA that could explain biological behavior. However, ploidy was higher in late-stage (median, 3.0) than early-stage (median, 1.9) samples. CN signature exposures were significantly different between cohorts, with greater relative signature 3 exposure in early-stage and greater signature 4 in late-stage. Unsupervised clustering based on CN signatures identified three clusters that were prognostic. Conclusions: Early-stage and late-stage HGSCs have highly similar patterns of mutation and focal SCNA. However, CN signature analysis showed that late-stage disease has distinct signature exposures consistent with whole-genome duplication. Further analyses will be required to ascertain whether these differences reflect genuine biological differences between early-stage and late-stage or simply time-related markers of evolutionary fitness. See related commentary by Yang et al., p. 2730
- Published
- 2021
27. Activating a collaborative innate-adaptive immune response to control metastasis
- Author
-
Xue-Yan He, David Ng, Xiao Han, Bodu Liu, Ana S. Almeida, Tim Kees, Phyllis A. Gimotty, Lijuan Sun, Iain A. McNeish, Sylvia Adams, David L. Spector, Mikala Egeblad, and Ovarian Cancer Action
- Subjects
Cancer Research ,Macrophage polarization ,Monophosphoryl Lipid A ,Metastasis ,Mice ,cytotoxic T cells ,Immune system ,breast cancer ,MPLA ,Animals ,Humans ,Medicine ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Neoplasm Metastasis ,anti-tumor immune response ,business.industry ,tumor-associated macrophages ,Macrophages ,Cancer ,medicine.disease ,Acquired immune system ,Immunity, Innate ,metastasis treatment ,ovarian cancer ,Oncology ,Cancer cell ,Cancer research ,Tumor necrosis factor alpha ,business ,1109 Neurosciences ,IFNγ - Abstract
Summary Tumor-associated macrophages (TAMs) promote metastasis and inhibit T cells, but macrophages can be polarized to kill cancer cells. Macrophage polarization could thus be a strategy for controlling cancer. We show that macrophages from metastatic pleural effusions of breast cancer patients can be polarized to kill cancer cells with monophosphoryl lipid A (MPLA) and interferon (IFN) γ. MPLA + IFNγ injected intratumorally or intraperitoneally reduces primary tumor growth and metastasis in breast cancer mouse models, suppresses metastasis, and enhances chemotherapy response in an ovarian cancer model. Both macrophages and T cells are critical for the treatment's anti-metastatic effects. MPLA + IFNγ stimulates type I IFN signaling, reprograms CD206+ TAMs to inducible NO synthase (iNOS)+ macrophages, and activates cytotoxic T cells through macrophage-secreted interleukin-12 (IL-12) and tumor necrosis factor alpha (TNFα). MPLA and IFNγ are used individually in clinical practice and together represent a previously unexplored approach for engaging a systemic anti-tumor immune response.
- Published
- 2021
28. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance through STING
- Author
-
Matteo Morotti, Sora Chee, Eleonora Ghisoni, Julien Dagher, Mauro Delorenzi, Raphael Genolet, George Coukos, David Barras, Lana E. Kandalaft, Denarda Dangaj Laniti, Michal Bassani-Sternberg, Catherine Ronet, Bing Ren, Julien Dorier, Alizée J. Grimm, Marco Mina, Alexandre Harari, Christian Iseli, Josephine Walton, Mathieu Desbuisson, Hualing Zhang, Theodora Tsianou, Sara Bobisse, Giovanni Ciriello, Melita Irving, Brian Stevenson, Sylvie Rusakiewicz, Elizabeth M. Swisher, Chloe Chong, Noémie Fahr, Evripidis Lanitis, Periklis G. Foukas, Iain A. McNeish, Fabio Martinon, Marine Bruand, Cancer Research UK, and Ovarian Cancer Action
- Subjects
Vascular Endothelial Growth Factor A ,Chemokine ,Transcription, Genetic ,T-Lymphocytes ,0601 Biochemistry and Cell Biology ,VEGF-A ,Epigenesis, Genetic ,angiogenesis ,Medicine ,dual immune checkpoint blockade ,Chemokine CCL5 ,Immune Checkpoint Inhibitors ,Ovarian Neoplasms ,biology ,Neovascularization, Pathologic ,BRCA1 Protein ,Chromatin ,ovarian cancer ,Female ,medicine.symptom ,DNA sensing ,PD-L1 ,PARPi ,T cells ,Inflammation ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,CCL5 ,Article ,Cell Line, Tumor ,Animals ,Humans ,Gene Silencing ,ICB ,business.industry ,Membrane Proteins ,DNA ,BRCA1 ,Immune checkpoint ,eye diseases ,Mice, Inbred C57BL ,Sting ,CTLA-4 ,Tumor progression ,1116 Medical Physiology ,biology.protein ,Cancer research ,Interferons ,Neoplasm Grading ,business ,type I IFN ,DNA Damage ,STING - Abstract
SUMMARY In this study, we investigate mechanisms leading to inflammation and immunoreactivity in ovarian tumors with homologous recombination deficiency (HRD). BRCA1 loss is found to lead to transcriptional reprogramming in tumor cells and cell-intrinsic inflammation involving type I interferon (IFN) and stimulator of IFN genes (STING). BRCA1-mutated (BRCA1mut) tumors are thus T cell inflamed at baseline. Genetic deletion or methylation of DNA-sensing/IFN genes or CCL5 chemokine is identified as a potential mechanism to attenuate T cell inflammation. Alternatively, in BRCA1mut cancers retaining inflammation, STING upregulates VEGF-A, mediating immune resistance and tumor progression. Tumor-intrinsic STING elimination reduces neoangiogenesis, increases CD8+ T cell infiltration, and reverts therapeutic resistance to dual immune checkpoint blockade (ICB). VEGF-A blockade phenocopies genetic STING loss and synergizes with ICB and/or poly(ADP-ribose) polymerase (PARP) inhibitors to control the outgrowth of Trp53−/−Brca1−/− but not Brca1+/+ ovarian tumors in vivo, offering rational combinatorial therapies for HRD cancers., Graphical abstract, In brief Bruand et al. provide insights into the dual role of STING in promoting tumor-intrinsic mechanisms of both immunoreactivity, driven by DNA sensing and type I IFN, and also VEGF-A-driven immune resistance in BRCA1mut ovarian cancers. STING elimination reduces neoangiogenesis, increases CD8+ T cell infiltration, and reverts therapeutic resistance to dual ICB.
- Published
- 2021
29. Inactivating mutations and X-ray crystal structure of the tumor suppressor OPCML reveal cancer-associated functions
- Author
-
Haonan Lu, Mohammad N. Alomary, Hani Gabra, Chiara Recchi, Evdoxia Karali, Edward W. Tate, Naomi E. Chayen, Manuela Mura, Zachary Maben, Grant C. Weaver, Aline T. Marinho, Mollie Jurewicz, James R. Birtley, Eloise V. N. Morecroft, Lawrence J. Stern, Jane Antony, Claudia von Arx, Elisa Zanini, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,BIOMARKER ,Glycosylation ,PROMOTER ,General Physics and Astronomy ,PROTEIN ,02 engineering and technology ,Crystallography, X-Ray ,medicine.disease_cause ,Receptor tyrosine kinase ,Epigenesis, Genetic ,law.invention ,law ,Missense mutation ,Tumour-suppressor proteins ,lcsh:Science ,Ovarian Neoplasms ,Mutation ,Multidisciplinary ,biology ,METHYLATION ,021001 nanoscience & nanotechnology ,Phenotype ,Cell invasion ,3. Good health ,Multidisciplinary Sciences ,INTERFACE ,Cell Transformation, Neoplastic ,Science & Technology - Other Topics ,Female ,0210 nano-technology ,EXPRESSION ,Tumor suppressor gene ,Science ,Mutation, Missense ,GPI-Linked Proteins ,NEURITE OUTGROWTH ,Protein Aggregation, Pathological ,Article ,General Biochemistry, Genetics and Molecular Biology ,OVARIAN-CANCER ,03 medical and health sciences ,Ovarian cancer ,DIGLONS ,MD Multidisciplinary ,medicine ,Humans ,Neoplasm Invasiveness ,X-ray crystallography ,Science & Technology ,Growth factor signalling ,Cancer ,General Chemistry ,medicine.disease ,GENE ,Protein Structure, Tertiary ,030104 developmental biology ,Cancer research ,biology.protein ,Suppressor ,lcsh:Q ,Carcinogenesis ,Cell Adhesion Molecules - Abstract
OPCML, a tumor suppressor gene, is frequently silenced epigenetically in ovarian and other cancers. Here we report, by analysis of databases of tumor sequences, the observation of OPCML somatic missense mutations from various tumor types and the impact of these mutations on OPCML function, by solving the X-ray crystal structure of this glycoprotein to 2.65 Å resolution. OPCML consists of an extended arrangement of three immunoglobulin-like domains and homodimerizes via a network of contacts between membrane-distal domains. We report the generation of a panel of OPCML variants with representative clinical mutations and demonstrate clear phenotypic effects in vitro and in vivo including changes to anchorage-independent growth, interaction with activated cognate receptor tyrosine kinases, cellular migration, invasion in vitro and tumor growth in vivo. Our results suggest that clinically occurring somatic missense mutations in OPCML have the potential to contribute to tumorigenesis in a variety of cancers., OPCML is a tumour suppressor gene that is epigenetically silenced in ovarian cancer and is somatically mutated in various cancers. Here, the authors solve the X-ray crystal structure of OPCML and model clinically relevant mutations that could contribute to tumorigenesis.
- Published
- 2019
30. Traditional Systemic Treatment Options in Advanced Low-Grade Serous Ovarian Cancer after Successful Cytoreduction: A Systematic Review and Meta-Analysis
- Author
-
Rosa Montero-Macías, Pascal Rigolet, Elie Mikhael, Jonathan Krell, Vincent Villefranque, Fabrice Lecuru, Christina Fotopoulou, and Ovarian Cancer Action
- Subjects
low-grade serous ovarian cancer ,Cancer Research ,Science & Technology ,hormonal therapy ,CARCINOMA ,PERITONEUM ,CLINICAL BEHAVIOR ,adjuvant treatment ,WOMEN ,CHEMOTHERAPY ,THERAPY ,Oncology ,SURVIVAL ,1112 Oncology and Carcinogenesis ,Life Sciences & Biomedicine - Abstract
Objective: We performed a systematic literature review and a subsequent meta-analysis to compare traditional treatment options, i.e., antihormonal and cytotoxic, in LGSOC. Methods: We conducted a systematic literature review in MEDBASE and MEDLINE between September 2000 and June 2021 for women who received cytotoxic chemotherapy and/or antihormonal treatment after primary cytoreduction due to stage II–IV LGSOC and also at relapse. PFS and OS were calculated depending on the type of their adjuvant treatment. For each endpoint in the meta-analysis, pooled HR was calculated using the random effect model with the inverse variance weighted method. Only primary patients were included in the subsequent meta-analysis due to the small number of studies in the relapsed setting. Results: Five eligible first-line studies were included. Systemic chemotherapy failed to provide a significant OS benefit when compared to no systemic treatment (pooled HR = 1.01, 95% CI [0.79, 1.29]) after successful cytoreduction. Moreover, systemic chemotherapy followed by antihormonal treatment also did not result to a significant PFS or OS benefit when compared to systemic chemotherapy alone (for PSF: pooled HR = 0.59, 95% CI [0.33, 1.04]; for OS: pooled HR = 0.83, 95% CI [0.50, 1.39]). There were insufficient data from studies in the recurrent setting to allow their inclusion in the meta-analysis. Conclusions: In this meta-analysis, we failed to identify a traditional cytotoxic or antihormonal systemic treatment option that was associated with a significant OS or PFS benefit when administered following successful cytoreduction for advanced LGSOC. Prospective randomized studies are urgently warranted to define optimal adjuvant options in this challenging disease.
- Published
- 2022
31. Chromatin accessibility changes at intergenic regions associate with ovarian cancer drug resistance
- Author
-
Erick Loomis, Ian M. Garner, Robert S. Brown, John Gallon, Leigh Brody, Edward Curry, James M. Flanagan, Nicholas Martin, Cancer Research UK, and Ovarian Cancer Action
- Subjects
Epigenomics ,DNA damage ,Antineoplastic Agents ,Drug resistance ,Biology ,Intergenic region ,Cell Line, Tumor ,Gene expression ,medicine ,Genetics ,Humans ,Chemotherapy ,Molecular Biology ,Gene ,Genetics (clinical) ,Cancer ,Ovarian Neoplasms ,0604 Genetics ,Research ,Promoter ,1103 Clinical Sciences ,DNA Methylation ,medicine.disease ,Chromatin ,Ovarian ,Drug Resistance, Neoplasm ,Cancer research ,1114 Paediatrics and Reproductive Medicine ,DNA, Intergenic ,Female ,Ovarian cancer ,Developmental Biology - Abstract
Background Resistance to DNA damaging chemotherapies leads to cancer treatment failure and poor patient prognosis. We investigated how genomic distribution of accessible chromatin sites is altered during acquisition of cisplatin resistance using matched ovarian cell lines from high grade serous ovarian cancer (HGSOC) patients before and after becoming clinically resistant to platinum-based chemotherapy. Results Resistant lines show altered chromatin accessibility at intergenic regions, but less so at gene promoters. Clusters of cis-regulatory elements at these intergenic regions show chromatin changes that are associated with altered expression of linked genes, with enrichment for genes involved in the Fanconi anemia/BRCA DNA damage response pathway. Further, genome-wide distribution of platinum adducts associates with the chromatin changes observed and distinguish sensitive from resistant lines. In the resistant line, we observe fewer adducts around gene promoters and more adducts at intergenic regions. Conclusions Chromatin changes at intergenic regulators of gene expression are associated with in vivo derived drug resistance and Pt-adduct distribution in patient-derived HGSOC drug resistance models.
- Published
- 2021
32. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2)
- Author
-
Ling Ma, Tanya T. Kwan, Clare L. Scott, Isabelle Ray-Coquard, Ana Oaknin, Alexander Dobrovic, Robert L. Coleman, Iain A. McNeish, Andrea E. Wahner Hendrickson, Lee-may Chen, Alexandra Leary, Stephen Welch, Thomas Harding, Lara Maloney, Carol Aghajanian, Kevin K. Lin, Heidi Giordano, E. Dominy, Ashan Musafer, Gottfried E. Konecny, Scott H. Kaufmann, Diane Provencher, Julia A. Elvin, Oliver Dorigo, Sandra Goble, R Kristeleit, Douglas I. Lin, Anna V. Tinker, Amit M. Oza, Setsuko K. Chambers, David M. O'Malley, Elizabeth M. Swisher, Prafull Ghatage, Lan Thanh Vo, Institut Català de la Salut, [Swisher EM] University of Washington, Seattle, WA, USA. [Kwan TT] Clovis Oncology, Inc., Boulder, CO, USA. [Oza AM] Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. [Tinker AV] BC Cancer—Vancouver, Vancouver, BC, Canada. [Ray-Coquard I] GINECO, Centre Léon Bérard and University Claude Bernard, Lyon, France. [Oaknin A] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Indoles ,endocrine system diseases ,General Physics and Astronomy ,neoplasias::neoplasias por localización::neoplasias de las glándulas endocrinas::neoplasias ováricas [ENFERMEDADES] ,Carcinoma, Ovarian Epithelial ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Poly (ADP-Ribose) Polymerase Inhibitor ,Tumour biomarkers ,chemistry.chemical_compound ,0302 clinical medicine ,Ovarian carcinoma ,Ovarian Epithelial ,80 and over ,Medicine ,Promoter Regions, Genetic ,Cancer ,Aged, 80 and over ,Ovarian Neoplasms ,neoplasias::procesos neoplásicos::recurrencia neoplásica local [ENFERMEDADES] ,Multidisciplinary ,BRCA1 Protein ,Ovaris - Càncer - Tractament ,Cell cycle ,Middle Aged ,BRCA2 Protein ,female genital diseases and pregnancy complications ,Ovarian Cancer ,DNA-Binding Proteins ,Local ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,RAD51C ,Female ,Adult ,Science ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,General Biochemistry, Genetics and Molecular Biology ,Article ,Promoter Regions ,03 medical and health sciences ,Rare Diseases ,Genetic ,Clinical Research ,Ovarian cancer ,Neoplasms::Neoplastic Processes::Neoplasm Recurrence, Local [DISEASES] ,Genetics ,Humans ,Rucaparib ,Ovaris - Càncer - Recaiguda ,Aged ,Platinum ,business.industry ,Neoplasms::Neoplasms by Site::Endocrine Gland Neoplasms::Ovarian Neoplasms [DISEASES] ,Carcinoma ,General Chemistry ,DNA Methylation ,030104 developmental biology ,Good Health and Well Being ,Neoplasm Recurrence ,chemistry ,Cancer research ,Neoplasm Recurrence, Local ,business - Abstract
ARIEL2 (NCT01891344) is a single-arm, open-label phase 2 study of the PARP inhibitor (PARPi) rucaparib in relapsed high-grade ovarian carcinoma. In this post hoc exploratory biomarker analysis of pre- and post-platinum ARIEL2 samples, RAD51C and RAD51D mutations and high-level BRCA1 promoter methylation predict response to rucaparib, similar to BRCA1/BRCA2 mutations. BRCA1 methylation loss may be a major cross-resistance mechanism to platinum and PARPi. Genomic scars associated with homologous recombination deficiency are irreversible, persisting even as platinum resistance develops, and therefore are predictive of rucaparib response only in platinum-sensitive disease. The RAS, AKT, and cell cycle pathways may be additional modulators of PARPi sensitivity., The identification of biomarkers of response to PARP inhibitors can enable selection of appropriate ovarian cancer patients for treatment. In this study, the authors report clinical results and exploratory biomarker analyses from the ARIEL2 phase 2 clinical trial on the safety and efficacy of the PARP inhibitor rucaparib in patients with recurrent ovarian cancers
- Published
- 2021
33. Population exposure-efficacy and exposure-safety analyses for rucaparib in patients with recurrent ovarian carcinoma from Study 10 and ARIEL2
- Author
-
Rebecca Kristeleit, Robert L. Coleman, Michelle Green, Anna V. Tinker, Amit M. Oza, Cesar M. Castro, James D. Brenton, David M. O'Malley, Ling Ma, Isabelle Ray-Coquard, Jim J. Xiao, Jeri Beltman, Diane Provencher, Thomas Harding, Kevin K. Lin, Elizabeth M. Swisher, Ana Oaknin, Ronnie Shapira-Frommer, Carol Aghajanian, Gottfried E. Konecny, Andrew Simmons, Sandra Goble, Alexandra Leary, Lara Maloney, Iain A. McNeish, Lee-may Chen, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Indoles ,Administration, Oral ,Carcinoma, Ovarian Epithelial ,Gastroenterology ,chemistry.chemical_compound ,0302 clinical medicine ,Ovarian Epithelial ,Medicine ,Ovarian carcinoma ,Cancer ,Ovarian Neoplasms ,education.field_of_study ,biology ,BRCA1 Protein ,Obstetrics and Gynecology ,Middle Aged ,Ovarian Cancer ,Oncology ,Local ,030220 oncology & carcinogenesis ,Area Under Curve ,6.1 Pharmaceuticals ,Administration ,Female ,Drug ,Safety ,Oral ,medicine.medical_specialty ,Efficacy ,Population ,Oncology and Carcinogenesis ,Cmax ,Aspartate transaminase ,Article ,Exposure ,Dose-Response Relationship ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Humans ,Pharmacokinetics ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,education ,Rucaparib ,Adverse effect ,Dose Modification ,Platinum ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Carcinoma ,Evaluation of treatments and therapeutic interventions ,030104 developmental biology ,Neoplasm Recurrence ,Orphan Drug ,Alanine transaminase ,chemistry ,biology.protein ,1114 Paediatrics and Reproductive Medicine ,Neoplasm Recurrence, Local ,business ,Recurrent Ovarian Carcinoma - Abstract
ObjectiveTo evaluate correlations between rucaparib exposure and selected efficacy and safety endpoints in patients with recurrent ovarian carcinoma using pooled data from Study 10 and ARIEL2.MethodsEfficacy analyses were limited to patients with carcinomas harboring a deleterious BRCA1 or BRCA2 mutation who had received ≥2 prior lines of chemotherapy. Safety was evaluated in all patients who received ≥1 rucaparib dose. Steady-state daily area under the concentration-time curve (AUCss) and maximum concentration (Cmax,ss) for rucaparib were calculated for each patient and averaged by actual dose received over time (AUCavg,ss and Cmax,avg,ss) using a previously developed population pharmacokinetic model.ResultsRucaparib exposure was dose-proportional and not associated with baseline patient weight. In the exposure-efficacy analyses (n=121), AUCavg,ss was positively associated with independent radiology review-assessed RECIST response in the subgroup of patients with platinum-sensitive recurrent disease (n=75, p=0.017). In the exposure-safety analyses (n=393, 40mg once daily to 840mg twice daily [BID] starting doses), most patients received a 600mg BID rucaparib starting dose, with 27% and 21% receiving 1 or≥2 dose reductions, respectively. Cmax,ss was significantly correlated with grade≥2 serum creatinine increase, grade≥3 alanine transaminase/aspartate transaminase increase, platelet decrease, fatigue/asthenia, and maximal hemoglobin decrease (p 
- Published
- 2021
34. FrenchFISH: Poisson Models for Quantifying DNA Copy Number From Fluorescence In Situ Hybridization of Tissue Sections
- Author
-
Edith M. Ross, Darren Ennis, Florian Markowetz, Teodora Goranova, David B. Morse, Ke Yuan, James D. Brenton, Jeremy A. Pike, Adam G. Berman, Anna M. Piskorz, Iain A. McNeish, Geoff Macintyre, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,DNA Copy Number Variations ,Tumor cells ,Poisson distribution ,01 natural sciences ,010104 statistics & probability ,03 medical and health sciences ,chemistry.chemical_compound ,symbols.namesake ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Computer Simulation ,Oncology & Carcinogenesis ,0101 mathematics ,In Situ Hybridization, Fluorescence ,Chromosome Aberrations ,medicine.diagnostic_test ,Cancer ,1103 Clinical Sciences ,General Medicine ,Gold standard (test) ,DNA ,medicine.disease ,Molecular biology ,030104 developmental biology ,Tissue sections ,chemistry ,symbols ,Fluorescence in situ hybridization - Abstract
PURPOSE Chromosomal aberration and DNA copy number change are robust hallmarks of cancer. The gold standard for detecting copy number changes in tumor cells is fluorescence in situ hybridization (FISH) using locus-specific probes that are imaged as fluorescent spots. However, spot counting often does not perform well on solid tumor tissue sections due to partially represented or overlapping nuclei. MATERIALS AND METHODS To overcome these challenges, we have developed a computational approach called FrenchFISH, which comprises a nuclear volume correction method coupled with two types of Poisson models: either a Poisson model for improved manual spot counting without the need for control probes or a homogeneous Poisson point process model for automated spot counting. RESULTS We benchmarked the performance of FrenchFISH against previous approaches using a controlled simulation scenario and tested it experimentally in 12 ovarian carcinoma FFPE-tissue sections for copy number alterations at three loci (c-Myc, hTERC, and SE7). FrenchFISH outperformed standard spot counting with 74% of the automated counts having < 1 copy number difference from the manual counts and 17% having < 2 copy number differences, while taking less than one third of the time of manual counting. CONCLUSION FrenchFISH is a general approach that can be used to enhance clinical diagnosis on sections of any tissue by both speeding up and improving the accuracy of spot count estimates.
- Published
- 2021
35. Metronomic oral cyclophosphamide in relapsed ovarian cancer
- Author
-
Rosalind Glasspool, Pavlina Spiliopoulou, Patricia Roxburgh, Samantha Hinsley, Iain A. McNeish, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
Oncology ,medicine.medical_treatment ,Administration, Oral ,PROGRESSION ,PACLITAXEL ,Carcinoma, Ovarian Epithelial ,THERAPY ,0302 clinical medicine ,Clinical endpoint ,Medicine ,RECURRENT ,TOPOTECAN ,Response rate (survey) ,Aged, 80 and over ,0303 health sciences ,education.field_of_study ,Obstetrics & Gynecology ,Obstetrics and Gynecology ,CHEMOTHERAPY ,Middle Aged ,PEGYLATED LIPOSOMAL DOXORUBICIN ,ovarian cancer ,030220 oncology & carcinogenesis ,Female ,BRCA1 protein ,Life Sciences & Biomedicine ,Adult ,medicine.medical_specialty ,Disease Response ,BEVACIZUMAB ,Population ,03 medical and health sciences ,LOW-DOSE CYCLOPHOSPHAMIDE ,Internal medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,education ,Adverse effect ,Cyclophosphamide ,030304 developmental biology ,Aged ,Retrospective Studies ,Chemotherapy ,Science & Technology ,business.industry ,medicine.disease ,RECIST ,BRCA2 protein ,Neoplasm Recurrence, Local ,business ,Ovarian cancer ,Progressive disease - Abstract
ObjectivesTo describe the clinical activity of metronomic cyclophosphamide in a population of patients with recurrent ovarian cancer, and to identify predictors of clinical response.MethodsWe retrospectively reviewed all patients treated at our institution with oral metronomic cyclophosphamide for relapsed ovarian cancer between January 2012 and December 2016. These were identified from electronic chemotherapy prescription records. The primary endpoint was response rate by combined Gynecologic Cancer InterGroup (GCIG) criteria. Data on patient demographics, previous therapies, platinum resistance, germline BRCA1/2 (gBRCA1/2) status, disease response by radiological or cancer antigen 125 (CA125) criteria alone, adverse events secondary to metronomic cyclophosphamide treatment, progression-free survival, and overall survival were also evaluated.Results50 out of 68 patients treated with oral metronomic cyclophosphamide were evaluable for disease response. By combination criteria (radiological plus CA125), complete response was 0%, partial response 32%, stable disease 16%, and progressive disease 52%. In the intention-to-treat population (n=68), progression-free survival and overall survival were 2.6 months and 6 months, respectively. Having a gBRCA1/2 mutation reduced the risk of disease progression by radiological criteria (OR 0.07, 95% CI 0.008 to 0.67, p=0.02), and patients with gBRCA1/2 mutations had improved progression-free survival (7.9 vs 2.5 months, HR 0.4, 95% CI 0.23 to 0.74, p=0.003) and overall survival (15.5 vs 6 months, HR 0.49, 95% CI 0.28 to 0.85, p=0.02) with metronomic cyclophosphamide when compared with patients without gBRCA1/2 mutations (or unknown gBRCA1/2 status).ConclusionOral metronomic cyclophosphamide showed a clinical benefit in 48% of patients with recurrent ovarian cancer. gBRCA1/2 status can be an independent predictor of response.
- Published
- 2021
36. The emerging role of Interleukin 1β (IL-1β) in cancer cachexia
- Author
-
Marie Fallon, Barry Laird, Donald C. McMillan, Iain J. Gallagher, Iain A. McNeish, D Robert Paval, Richard J E Skipworth, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Cachexia ,media_common.quotation_subject ,medicine.medical_treatment ,Interleukin-1beta ,Immunology ,Anti-Inflammatory Agents ,Adipose tissue ,Inflammation ,Review ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Immunology and Allergy ,Animals ,Humans ,Lung cancer ,media_common ,business.industry ,Interleukin-1 beta ,Appetite ,medicine.disease ,Canakinumab ,030104 developmental biology ,Cytokine ,1107 Immunology ,030220 oncology & carcinogenesis ,Cancer research ,Hypothalamic pituitary axis ,medicine.symptom ,Inflammation Mediators ,business ,Energy Metabolism ,medicine.drug - Abstract
Abstract Treatment of cancer cachexia remains an unmet need. The host-tumour interface and the resulting sequestration of the pro-inflammatory cytokine Il-1β is critical in cachexia development. Neuroinflammation mediated via IL-1β through the hypothalamic pituitary axis results in increased muscle proteolysis and adipose lipolysis, thus creating a prolonged stress-like environment with loss of appetite and increased resting energy expenditure. Recent trials using a monoclonal antibody targeting IL-1β, canakinumab, have shown a potential role in lung cancer; however, a potential role of targeting IL-1β to treat cachexia in patients with lung cancer is unclear, yet the underlying pathophysiology provides a sound rationale that this may be a viable therapeutic approach.
- Published
- 2021
37. Surgery due to mechanical bowel obstruction in relapsed ovarian cancer: clinical and surgical results of a bicentric analysis of 87 patients
- Author
-
Radoslav Chekerov, Christina Fotopoulou, Robert Armbrust, Jonathan Krell, S Sander, Natasha Rinne, Sascha Chopra, Jalid Sehouli, Katherine Nixon, Matthias Biebl, and Ovarian Cancer Action
- Subjects
Adult ,MAINTENANCE THERAPY ,medicine.medical_specialty ,Ileus ,medicine.medical_treatment ,Carcinoma, Ovarian Epithelial ,Stoma ,Young Adult ,medicine ,Humans ,Salvage surgery ,Obstetrics & Reproductive Medicine ,Aged ,Retrospective Studies ,Aged, 80 and over ,Ovarian Neoplasms ,Chemotherapy ,Science & Technology ,business.industry ,Obstetrics & Gynecology ,Obstetrics and Gynecology ,General Medicine ,Middle Aged ,medicine.disease ,Short bowel syndrome ,Surgery ,Bowel obstruction ,Serous fluid ,Parenteral nutrition ,1114 Paediatrics and Reproductive Medicine ,Neoplasm Recurrence, Local ,Recurrent ovarian cancer ,business ,Ovarian cancer ,Life Sciences & Biomedicine ,Intestinal Obstruction ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit::610 Medizin und Gesundheit - Abstract
Introduction Mechanical bowel obstruction is a frequent acute and life-threatening event in relapsed ovarian cancer. Salvage surgery after failure of all conservative approaches, resulting in short bowel syndrome (SBS) constitutes a therapeutic dilemma. Our aim was to evaluate patients’ surgical and clinical outcome in these highly palliative situations. Previous, limited, data reported a high morbidity and mortality. However, recent surgical and therapeutical improvements in relapsed ovarian cancer (ROC) offer better identification of patients who might benefit from surgery in an effort to extend the window of opportunity to subsequently offer these patients novel systemic therapeutic approaches. Material and methods All subsequent ROC patients between 2012 and 2017 with acute mechanical bowel obstruction who underwent salvage extraperitoneal en bloc intestinal resection were retrospectively identified. Data were collected from two ESGO certified Ovarian Cancer Centers of Excellence (Charité Berlin and Imperial College London) and systematically evaluated regarding surgical and clinical outcomes. Results Overall, 87 ROC patients were included in the analysis (median age 56 years, range 24–88), 47% were platinum resistant. High grade serous was the most common histology (76%) while most of the patients (67%) had at least two previous lines of treatment. Mean observed OS was 7.8 months. After salvage surgery, 46% of the patients had a residual small bowel length 180 cm resulting in 41% in need of total parental nutrition. In 80% of the patients a permanent stoma was necessary. 30d morbidity and mortality was 74% and 10%, respectively. More than half of the patients were able to receive further courses of chemotherapy after surgery. Discussion Salvage surgery for bowel obstruction in ROC patients needs careful consideration and identification of optimal surgical candidates to have the maximal therapeutic benefit. Despite the challenging morbidity profile, most patients managed to proceed to subsequent novel and conventional systemic treatment and so have their window of therapeutic opportunity extended.
- Published
- 2021
- Full Text
- View/download PDF
38. 805 Safety and emerging evidence of immune modulation of the live biotherapeutic MRx0518 in the neoadjuvant setting for patients awaiting surgical removal of solid tumours
- Author
-
Emily Pickford, Adam E Frampton, Alex Stevenson, Jonathan Krell, Axel Glasmacher, Mark P. Lythgoe, Marsilio Adriani, Gayle Fyvie, Justin Stebbing, Imperial College Healthcare NHS Trust- BRC Funding, National Institute for Health Research, 4D Pharma Plc, and Ovarian Cancer Action
- Subjects
Oncology ,medicine.medical_specialty ,medicine.diagnostic_test ,medicine.drug_class ,business.industry ,Melanoma ,medicine.medical_treatment ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunostimulant ,lcsh:RC254-282 ,Radiation therapy ,Immune system ,medicine.anatomical_structure ,Tolerability ,Prostate ,Internal medicine ,Biopsy ,medicine ,Adverse effect ,business - Abstract
Background The gut microbiome has emerged as a promising innovative therapeutic target for immune-stimulation treatment of solid tumours. MRx0518 is a novel, gut microbiome-derived oral live biotherapeutic. It has potent anti-tumorigenic efficacy in the preclinical setting including murine models of lung (LLC1), kidney (Renca) and breast (EMT6) cancer.1 In these models, a significant reduction in tumour growth has been demonstrated, including induction of immunostimulatory responses with tumour infiltration of NK cells, CD8+ and CD4+ T-cells. MRx0518 is under investigation in various oncological settings, including in combination with immune checkpoint inhibitors (NCT03637803) and radiotherapy (NCT04193904). Methods Treatment naive patients were recruited from April 2019 to February 2020. Patients were eligible if they received a histologically confirmed diagnosis of cancer (solid tumours) scheduled for surgical resection. Patients received 1 capsule of MRx0518 (1x1010 to 1x1011 CFU) twice daily from inclusion until the day preceding surgery (maximum 28 days therapy). The primary study outcome is to evaluate safety and tolerability of MRx0518 monotherapy in treatment naive patients. Additional exploratory outcomes including identifying surrogate biomarkers of efficacy, microbiome analysis, effect on metabonomic markers and identification of histological and genomic alterations in paired pre-treatment (diagnostic biopsy) and post-treatment (surgical specimen) samples. Results In part A, 17 patients received treatment, across tumour groups including breast (n=8), prostate (n=4), uterine (n=3), melanoma (n=1) and bladder (n=1). MRx0518 was well tolerated by all, with no grade 3/4 CTCAE toxicity reported, no severe adverse effects or treatment discontinuations. All patients proceeded to surgery, however the COVID-19 pandemic delayed surgery in 3 cases. Analysis of the first 5* patient paired samples utilising the NanoString Pan Cancer IO 360TM Gene Expression panel has demonstrated significant changes in gene expression profiles in 48 genes (p Conclusions This study has demonstrated the safety and tolerability of the live biotherapeutic MRx0518 in treatment naive cancer patients. Exploratory analyses of post-treatment samples has echoed preclinical observations of increased infiltration of immune cells following treatment and will undergo further validation. Part B will focus on investigating efficacy in a further 100 treatment naive patients with a placebo-controlled arm. Trial Registration NCT03934827 Ethics Approval The study was approved by East of England - Cambridge East Research Ethics Committee approval number 18/EE/0091 Reference Laute-Caly DL, Raftis EJ, Cowie P, et al. The flagellin of candidate live biotherapeutic Enterococcus gallinarum MRx0518 is a potent immunostimulant. Scientific Reports 2019;9(1):1-14. doi:10.1038/s41598-018-36926-8 *Data analysis has been censored at 18/9/2020, further samples analysis is ongoing and will be updated.
- Published
- 2020
39. Glomerular filtration rate estimation for carboplatin dosing in patients with gynaecological cancers
- Author
-
A. Samani, R. Bennett, K. Eremeishvili, F. Kalofonou, S. Whear, A. Montes, R. Kristeleit, J. Krell, I. McNeish, S. Ghosh, L. Tookman, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and National Institute for Health Research
- Subjects
glomerular filtration rate ,Cancer Research ,Oncology ,gynaecological cancers ,carboplatin ,toxicity ,urologic and male genital diseases ,chemotherapy ,female genital diseases and pregnancy complications - Abstract
Background: Carboplatin remains integral for treatment of gynaecological malignancies and dosing is based on glomerular filtration rate (GFR). Measurement via radiotracer decay (nmGFR) is ideal. However, this may be unavailable. Therefore, GFR is often estimated using formulae that have not been validated in patients with cancer and/or specifically for gynaecological malignancies, leading to debate over optimal estimation. Suboptimal GFR estimation may affect efficacy or toxicity. Methods: We surveyed several UK National Health Service Trusts to assess carboplatin dosing practise. We then explored single-centre accuracy, bias and precision of various formulae for GFR estimation, relative to nmGFR, before validating our findings in an external cohort. Results: Across 18 Trusts, there was considerable heterogeneity in GFR estimation, including the formulae used (Cockcroft-Gault (CG) vs Wright), weight-adjustment and area under the curve (5 vs 6). We analysed 274 and 192 patients in two centres. Overall, CamGFR v2 (a novel formula for GFR estimation developed at Cambridge University Hospitals NHS Foundation Trust) excelled, showing the highest accuracy and precision. This translated into accuracy of hypothetical carboplatin dosing; nmGFR-derived carboplatin dose fell within 20% of the Cam GFR v2-derived dose in 86.5% and 87% of patients across the cohorts. Amongst the CG formula and its derivatives, using adjusted body weight in those with BMI ≥25 kg/m2 (CG-AdBW) was optimal. The Wright and unadjusted CG estimators performed most poorly. Conclusions: When compared with nmGFR assessment, accuracy, bias and precision varied widely between GFR estimators, with the newly developed Cam GFR v2 and CG-AdBW perfoming best. In general, weight (or body surface area)-adjusted formulae performed best, while the unadjusted CG and Wright formulae or the use of AUC6 (vs. nmGFR AUC5) produced risk of significant overdose. Thus, individual centres should validate their GFR estimation methods. In the absence of validation, CG-AdBW or CamGFR v2 are likely to perform well while unadjusted CG /Wright formulae or AUC6 dosing should be avoided.
- Published
- 2022
40. The Clinical Significance of Transfer RNAs Present in Extracellular Vesicles
- Author
-
Daniel S. K. Liu, Qi Zhi Clayton Yang, Mohammad Asim, Jonathan Krell, Adam E. Frampton, Ovarian Cancer Action, and Curesponse Ltd.
- Subjects
EXPRESSION ,Biochemistry & Molecular Biology ,Chemistry, Multidisciplinary ,BIOMARKERS ,0699 Other Biological Sciences ,Exosomes ,Catalysis ,Inorganic Chemistry ,Extracellular Vesicles ,RNA, Transfer ,DIAGNOSTIC-VALUE ,0399 Other Chemical Sciences ,exosome ,cancer ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,FRAGMENTS ,Spectroscopy ,tRNA fragment ,0604 Genetics ,Science & Technology ,Chemical Physics ,tRNA half ,Organic Chemistry ,General Medicine ,transfer RNA ,Computer Science Applications ,Chemistry ,NONCODING RNAS ,EXOSOMAL MIRNAS ,Physical Sciences ,CELLS ,extracellular vesicle ,Life Sciences & Biomedicine - Abstract
Extracellular vesicles (EVs) are important for intercellular signalling in multi-cellular organisms. However, the role of mature transfer RNAs (tRNAs) and tRNA fragments in EVs has yet to be characterised. This systematic review aimed to identify up-to-date literature on tRNAs present within human EVs and explores their potential clinical significance in health and disease. A comprehensive and systematic literature search was performed, and the study was conducted in accordance with PRISMA guidelines. Electronic databases MEDLINE and EMBASE were searched up until 1 January 2022. From 685 papers, 60 studies were identified for analysis. The majority of papers reviewed focussed on the role of EV tRNAs in cancers (31.7%), with numerous other conditions represented. Blood and cell lines were the most common EV sources, representing 85.9% of protocols used. EV isolation methods included most known methods, precipitation being the most common (49.3%). The proportion of EV tRNAs was highly variable, ranging between 0.04% to >95% depending on tissue source. EV tRNAs are present in a multitude of sources and show promise as disease markers in breast cancer, gastrointestinal cancers, and other diseases. EV tRNA research is an emerging field, with increasing numbers of papers highlighting novel methodologies for tRNA and tRNA fragment discovery.
- Published
- 2022
41. Race reporting and diversity in US food and drug administration (FDA) registration trials for prostate cancer; 2006-2020
- Author
-
Jonathan Krell, Mark P. Lythgoe, Philip Savage, Vinay Prasad, and Ovarian Cancer Action
- Subjects
Urologic Diseases ,Male ,Cancer Research ,medicine.medical_specialty ,Aging ,Urology ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,030232 urology & nephrology ,03 medical and health sciences ,Prostate cancer ,Race (biology) ,0302 clinical medicine ,Clinical Research ,Epidemiology of cancer ,Medicine ,Humans ,1112 Oncology and Carcinogenesis ,Disease burden ,American Indian or Alaska Native ,Cancer ,Retrospective Studies ,Clinical Trials as Topic ,Science & Technology ,business.industry ,United States Food and Drug Administration ,Incidence (epidemiology) ,Prevention ,Prostate Cancer ,Incidence ,Evaluation of treatments and therapeutic interventions ,Prostatic Neoplasms ,Urology & Nephrology ,medicine.disease ,United States ,Clinical trial ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,6.1 Pharmaceuticals ,Pacific islanders ,Outcomes research ,business ,Life Sciences & Biomedicine - Abstract
Background There is significant racial disparity in prostate cancer (PCa) in terms of incidence, treatment, and outcomes. Racial diversity and compliance with FDA race reporting guidelines in PCa drug registration trials are unknown. We analyzed racial diversity and race reporting in drug licensing trials for PCa. Methods New drug authorizations for PCa from 2006 to 2020 were identified. The corresponding licensing trial publications were analyzed to check compliance with current FDA recommendations for race reporting. If race was unreported, the clinical trial report was analyzed to determine participant recruitment by race and lead the recruiting country. Results During the study period, 17 new drug registrations for the management of PCa involving ten unique drugs were identified. In total, 18,455 participants were included in FDA registration trials, of which 76.3% were white or Caucasian, 7.9% Asian, 2.9% Black or African American, 0.5% American Indian or Alaskan Native, 0.1% Native Hawaiian or other Pacific Islander, 1.8% other or multiple races and 10.5% unknown. 53% of trials reported race in the licensing publication, however of this only 55% met current FDA recommendations. When the race was unreported in the licensing publication, 88% of studies had further information in the clinical study report. Conclusion We found a significant under-representation of non-white participants in FDA drug registration trials for PCa. Race reporting in licensing publication is inconsistent and both FDA and International Committee of Medical Journal Editors guidelines are not being universally followed. Given the disproportionality of the disease burden of PCa, recruitment of Black and other minority participants to trials should be a research priority.
- Published
- 2020
42. An exploratory analysis of objective responses to neoadjuvant chemotherapy: results from a randomised phase III trial evaluating first-line carboplatin-paclitaxel regimens for ovarian, fallopian tube or primary peritoneal carcinoma (ICON8)
- Author
-
McNeish, I, Morgan, RD, Cook, AD, James, EC, Lord, R, Dark, G, Glasspool, RM, Krell, J, Parkinson, C, Poole, CJ, Hall, M, Gallardo-Rincón, D, Lockley, M, Essapen, S, Summers, J, Anand, A, Zachariah, A, Williams, S, Jones, R, Scatchard, K, Walther, A, Kim, J-W, Sundar, S, Jayson, GC, Ledermann, JA, Clamp, AR, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, and Cancer Research UK
- Subjects
1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
BackgroundPlatinum-based neoadjuvant chemotherapy (NACT) followed by delayed primary surgery (DPS) is an established strategy for women with newly diagnosed, advanced stage epithelial ovarian cancer. Although this therapeutic approach has been validated in randomised, phase III trials,evaluation of response to NACT using Response Evaluation Criteria in Solid Tumours (RECIST)and CA125 was not reported. We describeRECIST and Gynecologic Cancer InterGroup (GCIG)CA125 responses in patients receiving platinum-based NACT followed by DPS in the phase III trial, ICON8.MethodsICON8 was an international, multicentre, randomised, phase III trial in which women ≥18 years old with an Eastern Cooperative Oncology Group performance status of 0-2, life expectancy >12 weeks and newly diagnosed International Federation of Gynecology and Obstetrics (FIGO;1988) stage IC-IIA high-grade serous, clear cell or any poorly differentiated/grade 3 histological subtype or any FIGO (1988) stage IIB-IV epithelial cancer of the ovary, fallopian tube or primary peritoneum were randomised (1:1:1) to receive either intravenous (IV) carboplatin (AUC5/6)and IV paclitaxel (175mg/m2by body surface area [BSA])on day 1 of every 21-day cycle(control arm)or IV carboplatin (AUC5/6)on day 1 and IV paclitaxel (80mg/m2by BSA)on days 1, 8 and 15 of every 21-day cycle(dose-fractionated paclitaxelarm) or IV carboplatin (AUC2)and IV paclitaxel (80mg/m2by BSA)on days 1, 8 and 15 of every 21-day cycle(dose-fractionated carboplatin and paclitaxelarm). Randomisation occurred using a minimisation method and patients where stratified according to GCIG group, disease stage and timing and outcome of cytoreductive surgery. Neither patients nor clinicians were masked to their allocated group. The scheduling of surgery and use of NACTwere determined by local multidisciplinary case review. In this post-hoc 5exploratory analysis of ICON8, progression-free survival (PFS) was analysed using the landmark method and defined as the time interval between the date of pre-surgical planning radiological tumour assessment to the date of investigator-assessed clinical or radiological progression or death, whichever occurred first. This is different to the intention-to-treat primary PFS efficacy analysis of ICON8, which definedPFS as the time from randomisation to the date of clinical or radiological progression or death, whichever occurred first. This post-hoc exploratory analysis includes only women recruited to ICON8thatwere planned for NACT followed by DPS and had RECIST v1.1 and/or GCIG CA125 evaluable disease. ICON8isclosed for enrolment and follow-up, and registered with ClinicalTrials.gov, number: NCT01654146. Findings: BetweenJune 6,2011 and November 28,2014,1,566 women were enrolled in ICON8.Seven hundred and seventy-nine women were planned forNACT followed by DPS(NACT-DPS). In the NACT-DPS population,94% had FIGO stage IIIC/IV disease. Five hundred and sixty-four women had RECIST evaluable disease at trial entry and the complete or partial response rate (CR/PR) was 62%(348/564). Seven hundred and twenty-seven women were evaluable by GCIG CA125 criteria at the time of diagnosis and 84%(610/727) had a CA125 response. The median PFS was 14.4 months (95% CI [confidence interval] 9.2-28.0months; 297 events) for RECIST CR/PR and 13.3 months (95% CI 8.1-20.1months; 171 events) for RECIST stable disease(SD). The median PFS for those women with a GCIG CA125 response was 13.8 months (95% CI 8.8-23.4months; 544 events) and 9.7 months (95% CI 5.8-14.5months; 111 events) for those without. Complete cytoreduction(R0) was achieved in 56% (187/335) of women with RECISTCR/PRand42% (73/172) with RECIST SD. Complete cytoreduction (R0) was achieved in50% (290/576) and 30% (30/101) of women 6with and without a GCIG CA125 response, respectively. The median follow-up was 29.5 months (interquartile range:15.6-54.3months) for the NACT-DPS population.InterpretationThe RECIST-defined radiological response was lower than frequently quoted to patients in the clinic. RECIST v1.1 and GCIGCA125 responses to NACT for epithelial ovarian cancer should not be used as individual predictive markers to stratify patients likely to benefit from DPS, but instead used in conjunction with the patient’s clinical capacity to undergo cytoreductive surgery. A patient should not be denied surgery based solely on the lack of a RECIST v1.1 or GCIG CA125 response.
- Published
- 2020
43. Objective responses to first-line neoadjuvant carboplatin-paclitaxel regimens for ovarian, fallopian tube, or primary peritoneal carcinoma (ICON8): post-hoc exploratory analysis of a randomised, phase 3 trial
- Author
-
Andrew R Clamp, Sharadah Essapen, Adrian Cook, Marcia Hall, Elizabeth C. James, Sarah Williams, Jonathan A. Ledermann, Kate Scatchard, Jae Weon Kim, Jonathan Krell, Abel Zachariah, Gordon C Jayson, Iain A. McNeish, Axel Walther, Anjana Anand, Rachel Jones, Sudha Sundar, Christine Parkinson, Jeff Summers, Robert D. Morgan, Dolores Gallardo-Rincón, Christopher J. Poole, Graham Dark, Rosemary Lord, Rosalind Glasspool, Michelle Lockley, Medical Research Council (MRC), Ovarian Cancer Action, Imperial College Healthcare NHS Trust- BRC Funding, and Cancer Research UK
- Subjects
Oncology ,medicine.medical_specialty ,Paclitaxel ,Population ,Disease-Free Survival ,law.invention ,Carboplatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Primary peritoneal carcinoma ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Fallopian Tube Neoplasms ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,030212 general & internal medicine ,education ,Survival analysis ,Fallopian Tubes ,Peritoneal Neoplasms ,Response Evaluation Criteria in Solid Tumors ,Aged ,Body surface area ,Ovarian Neoplasms ,education.field_of_study ,business.industry ,Australia ,Membrane Proteins ,Middle Aged ,medicine.disease ,Neoadjuvant Therapy ,Clinical trial ,chemistry ,030220 oncology & carcinogenesis ,CA-125 Antigen ,Female ,business ,Ireland ,New Zealand - Abstract
Summary Background Platinum-based neoadjuvant chemotherapy followed by delayed primary surgery (DPS) is an established strategy for women with newly diagnosed, advanced-stage epithelial ovarian cancer. Although this therapeutic approach has been validated in randomised, phase 3 trials, evaluation of response to neoadjuvant chemotherapy using Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST), and cancer antigen 125 (CA125) has not been reported. We describe RECIST and Gynecologic Cancer InterGroup (GCIG) CA125 responses in patients receiving platinum-based neoadjuvant chemotherapy followed by DPS in the ICON8 trial. Methods ICON8 was an international, multicentre, randomised, phase 3 trial done across 117 hospitals in the UK, Australia, New Zealand, Mexico, South Korea, and Ireland. The trial included women aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0–2, life expectancy of more than 12 weeks, and newly diagnosed International Federation of Gynecology and Obstetrics (FIGO; 1988) stage IC–IIA high-grade serous, clear cell, or any poorly differentiated or grade 3 histological subtype, or any FIGO (1988) stage IIB–IV epithelial cancer of the ovary, fallopian tube, or primary peritoneum. Patients were randomly assigned (1:1:1) to receive intravenous carboplatin (area under the curve [AUC]5 or AUC6) and intravenous paclitaxel (175 mg/m2 by body surface area) on day 1 of every 21-day cycle (control group; group 1); intravenous carboplatin (AUC5 or AUC6) on day 1 and intravenous dose-fractionated paclitaxel (80 mg/m2 by body surface area) on days 1, 8, and 15 of every 21-day cycle (group 2); or intravenous dose-fractionated carboplatin (AUC2) and intravenous dose-fractionated paclitaxel (80 mg/m2 by body surface area) on days 1, 8, and 15 of every 21-day cycle (group 3). The maximum number of cycles of chemotherapy permitted was six. Randomisation was done with a minimisation method, and patients were stratified according to GCIG group, disease stage, and timing and outcome of cytoreductive surgery. Patients and clinicians were not masked to group allocation. The scheduling of surgery and use of neoadjuvant chemotherapy were determined by local multidisciplinary case review. In this post-hoc exploratory analysis of ICON8, progression-free survival was analysed using the landmark method and defined as the time interval between the date of pre-surgical planning radiological tumour assessment to the date of investigator-assessed clinical or radiological progression or death, whichever occurred first. This definition is different from the intention-to-treat primary progression-free survival analysis of ICON8, which defined progression-free survival as the time from randomisation to the date of first clinical or radiological progression or death, whichever occurred first. We also compared the extent of surgical cytoreduction with RECIST and GCIG CA125 responses. This post-hoc exploratory analysis includes only women recruited to ICON8 who were planned for neoadjuvant chemotherapy followed by DPS and had RECIST and/or GCIG CA125-evaluable disease. ICON8 is closed for enrolment and follow-up, and registered with ClinicalTrials.gov , NCT01654146 . Findings Between June 6, 2011, and Nov 28, 2014, 1566 women were enrolled in ICON8, of whom 779 (50%) were planned for neoadjuvant chemotherapy followed by DPS. Median follow-up was 29·5 months (IQR 15·6–54·3) for the neoadjuvant chemotherapy followed by DPS population. Of 564 women who had RECIST-evaluable disease at trial entry, 348 (62%) had a complete or partial response. Of 727 women who were evaluable by GCIG CA125 criteria at the time of diagnosis, 610 (84%) had a CA125 response. Median progression-free survival was 14·4 months (95% CI 9·2–28·0; 297 events) for patients with a RECIST complete or partial response and 13·3 months (8·1–20·1; 171 events) for those with RECIST stable disease. Median progression-free survival for women with a GCIG CA125 response was 13·8 months (95% CI 8·8–23·4; 544 events) and 9·7 months (5·8–14·5; 111 events) for those without a GCIG CA125 response. Complete cytoreduction (R0) was achieved in 187 (56%) of 335 women with a RECIST complete or partial response and 73 (42%) of 172 women with RECIST stable disease. Complete cytoreduction was achieved in 290 (50%) of 576 women with a GCIG CA125 response and 30 (30%) of 101 women without a GCIG CA125 response. Interpretation The RECIST-defined radiological response rate was lower than that frequently quoted to patients in the clinic. RECIST and GCIG CA125 responses to neoadjuvant chemotherapy for epithelial ovarian cancer should not be used as individual predictive markers to stratify patients who are likely to benefit from DPS, but instead used in conjunction with the patient's clinical capacity to undergo cytoreductive surgery. A patient should not be denied surgery based solely on the lack of a RECIST or GCIG CA125 response. Funding Cancer Research UK, UK Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, and Cancer Australia.
- Published
- 2020
44. Detecting endometrial cancer by blood spectroscopy: A diagnostic cross-sectional study
- Author
-
Rhona J McVey, Kássio M. G. Lima, Helena O'Flynn, Emma J Crosbie, Sarah Kitson, Abigail E. Derbyshire, Cecilia Pow, Pierre L. Martin-Hirsch, Maria Paraskevaidi, Olivia Raglan, Katherine M. Ashton, Vanitha N Sivalingam, Helen F. Stringfellow, Camilo L. M. Morais, Maria Kyrgiou, Neil A J Ryan, Francis Martin, Michelle L. MacKintosh, Ovarian Cancer Action, and HCA International Limited
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Cross-sectional study ,Atypical hyperplasia ,SERUM ,0302 clinical medicine ,blood diagnostics ,Informed consent ,Blood plasma ,METABOLIC SYNDROME ,RISK ,0303 health sciences ,PLASMA ,medicine.diagnostic_test ,Manchester Cancer Research Centre ,Obstetrics ,Incidence (epidemiology) ,WOMEN ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030220 oncology & carcinogenesis ,endometrial cancer ,Life Sciences & Biomedicine ,medicine.medical_specialty ,spectroscopy ,BIOMARKERS ,lcsh:RC254-282 ,Article ,CLASSIFICATION ,03 medical and health sciences ,Internal medicine ,medicine ,Blood test ,1112 Oncology and Carcinogenesis ,030304 developmental biology ,Research ethics ,Science & Technology ,Cancer prevention ,business.industry ,Endometrial cancer ,screening ,ResearchInstitutes_Networks_Beacons/mcrc ,B230 ,Cancer ,medicine.disease ,TRENDS ,030104 developmental biology ,business - Abstract
Endometrial cancer is the sixth most common cancer in women, with a rising incidence worldwide. Current approaches for the diagnosis and screening of endometrial cancer are invasive, expensive or of moderate diagnostic accuracy, limiting their clinical utility. There is a need for cost-effective and minimally invasive approaches to facilitate the early detection and timely management of endometrial cancer. We analysed blood plasma samples in a cross-sectional diagnostic accuracy study of women with endometrial cancer (n = 342), its precursor lesion atypical hyperplasia (n = 68) and healthy controls (n = 242, total n = 652) using attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy and machine learning algorithms. We show that blood-based infrared spectroscopy has the potential to detect endometrial cancer with 87% sensitivity and 78% specificity. Its accuracy is highest for Type I endometrial cancer, the most common subtype, and for atypical hyperplasia, with sensitivities of 91% and 100%, and specificities of 81% and 88%, respectively. Our large-cohort study shows that a simple blood test could enable the early detection of endometrial cancer of all stages in symptomatic women and provide the basis of a screening tool in high-risk groups. Such a test has the potential not only to differentially diagnose endometrial cancer but also to detect its precursor lesion atypical hyperplasia&mdash, the early recognition of which may allow fertility sparing management and cancer prevention.
- Published
- 2020
45. Paclitaxel induces immunogenic cell death in ovarian cancer via TLR4/IKK2/SNARE-dependent exocytosis
- Author
-
Tat San Lau, Chi Hang Wong, Loucia K.Y. Chan, Joseph Kwong, Tak Hong Cheung, So Fan Yim, Gene Chi Wai Man, Iain A. McNeish, Jacqueline Ho Sze Lee, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Adult ,Cancer Research ,Paclitaxel ,medicine.medical_treatment ,Immunology ,Cell ,Antineoplastic Agents ,Immunogenic Cell Death ,Cancer Vaccines ,Exocytosis ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,1112 Oncology and Carcinogenesis ,Aged ,Ovarian Neoplasms ,Chemotherapy ,business.industry ,Kinase ,Middle Aged ,medicine.disease ,I-kappa B Kinase ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,1107 Immunology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Immunogenic cell death ,Female ,Signal transduction ,1115 Pharmacology and Pharmaceutical Sciences ,business ,Ovarian cancer ,SNARE Proteins ,Signal Transduction - Abstract
Emerging evidence shows that the efficacy of chemotherapeutic drugs is reliant on their capability to induce immunogenic cell death (ICD), thus transforming dying tumor cells into antitumor vaccines. We wanted to uncover potential therapeutic strategies that target ovarian cancer by having a better understanding of the standard-of-care chemotherapy treatment. Here, we showed in ovarian cancer that paclitaxel induced ICD-associated damage-associated molecular patterns (DAMP, such as CALR exposure, ATP secretion, and HMGB1 release) in vitro and elicited significant antitumor responses in tumor vaccination assays in vivo. Paclitaxel-induced TLR4 signaling was essential to the release of DAMPs, which led to the activation of NF-κB–mediated CCL2 transcription and IkappaB kinase 2–mediated SNARE-dependent vesicle exocytosis, thus exposing CALR on the cell surface. Paclitaxel induced endoplasmic reticulum stress, which triggered protein kinase R–like ER kinase activation and eukaryotic translation initiation factor 2α phosphorylation independent of TLR4. Paclitaxel chemotherapy induced T-cell infiltration in ovarian tumors of the responsive patients; CALR expression in primary ovarian tumors also correlated with patients' survival and patient response to chemotherapy. These findings suggest that the effectiveness of paclitaxel relied upon the activation of antitumor immunity through ICD via TLR4 and highlighted the importance of CALR expression in cancer cells as an indicator of response to paclitaxel chemotherapy in ovarian cancer.
- Published
- 2020
46. Oncologist-led BRCA ‘mainstreaming’ in the ovarian cancer clinic: A study of 255 patients and its impact on their management
- Author
-
Iain A. McNeish, Nazneen Rahman, Hani Gabra, Megan Rumford, Angela George, Mark P. Lythgoe, Jonathan Krell, Laura A. Tookman, Imperial College Healthcare NHS Trust- BRC Funding, Biomedical Research Council, and Ovarian Cancer Action
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,endocrine system diseases ,medicine.medical_treatment ,Decision Making ,lcsh:Medicine ,Poly(ADP-ribose) Polymerase Inhibitors ,Mainstreaming ,Article ,Germline ,Germline mutation ,Ovarian cancer ,Internal medicine ,medicine ,Humans ,lcsh:Science ,Germ-Line Mutation ,Aged ,Platinum ,Aged, 80 and over ,BRCA2 Protein ,Ovarian Neoplasms ,Chemotherapy ,Multidisciplinary ,Molecular medicine ,BRCA1 Protein ,business.industry ,lcsh:R ,BRCA mutation ,Middle Aged ,Prognosis ,medicine.disease ,United Kingdom ,female genital diseases and pregnancy complications ,Cohort ,PARP inhibitor ,Female ,lcsh:Q ,business - Abstract
Although guidelines recommend BRCA testing for all women with non-mucinous epithelial ovarian cancer, there is significant variability in access to testing across the UK. A germline BRCA mutation (BRCAm) in ovarian cancer patients provides prognostic and predictive information and influences clinical management, such as the use of PARP inhibitors, which have demonstrated a progression-free survival benefit in the BRCAm cohort. Additionally, the finding of a BRCAm has significant implications for patients and their families in terms of cancer risk and prevention. We studied the impact of a newly-formed, oncologist-led ‘mainstreaming’ germline BRCA testing pathway in 255 ovarian cancer patients at Imperial College NHS Trust. Prior to the establishment of ‘mainstreaming’, uptake of germline BRCA testing was 14% with a mean turnaround time of 148.2 calendar days. The ‘mainstreaming’ approach led to a 95% uptake of germline BRCA testing and a mean turnaround time of 20.6 days. Thirty-four (13.33%) BRCAm patients were identified. At the time of data collection nine BRCAm patients had received a PARP inhibitor off-trial, three had entered a PARP inhibitor trial and 5 were receiving platinum-based chemotherapy with a plan to receive PARP inhibitor maintenance. This study provides further evidence of the impact of oncologist-led ‘mainstreaming’ programs.
- Published
- 2020
47. NK cells augment oncolytic adenovirus cytotoxicity in ovarian cancer
- Author
-
Jaya Nautiyal, Daniel M. Davis, Iain A. McNeish, Malcolm Farquharson, Sophie McNamara, Kerry D. Fisher, Christopher A.H. Hansell, Darren Ennis, Elaine Y.L. Leung, Philippa R. Kennedy, Suzanne Dowson, Leo M. Carlin, Pavlina Spiliopoulou, Gerard J. Graham, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, and Ovarian Cancer Action
- Subjects
0301 basic medicine ,Oncolytic adenovirus ,Cancer Research ,Enadenotucirev ,Oncolytic virus ,TIGIT ,T cell ,viruses ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Ovarian cancer ,medicine ,Adenovirus ,Pharmacology (medical) ,NK cell ,Cytotoxicity ,DNAM-1 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine - Abstract
Oncolytic viruses (OVs) can trigger profound innate and adaptive immune responses, which have the potential both to potentiate and reduce the activity of OVs. Natural killer (NK) cells can mediate potent anti-viral and anti-tumoral responses, but there are no data on the role of NK cells in oncolytic adenovirus activity. Here, we have used two different oncolytic adenoviruses—the Ad5 E1A CR2-deletion mutant dl922-947 (group C) and the chimeric Ad3/Ad11p mutant enadenotucirev (group B)—to investigate the effect of NK cells on overall anti-cancer efficacy in ovarian cancer. Because human adenoviruses do not replicate in murine cells, we utilized primary human NK cells from peripheral blood and ovarian cancer ascites. Our results show that dl922-947 and enadenotucirev do not infect NK cells, but induce contact-dependent activation and anti-cancer cytotoxicity against adenovirus-infected ovarian cancer cells. Moreover, manipulation of NK receptors DNAM-1 (DNAX accessory molecule-1) and TIGIT (T cell immunoreceptor with Ig and ITIM domains) significantly influences NK cytotoxicity against adenovirus-infected cells. Together, these results indicate that NK cells act to increase the activity of oncolytic adenovirus in ovarian cancer and suggest that strategies to augment NK activity further via the blockade of inhibitory NK receptor TIGIT could enhance therapeutic potential of OVs. Keywords: Ovarian cancer, Oncolytic virus, Adenovirus, NK cell, DNAM-1, TIGIT
- Published
- 2020
48. Tumors defective in homologous recombination rely on oxidative metabolism: relevance to treatments with PARP inhibitors
- Author
-
Alberto Villanueva, Roger Moreno, Iain A. McNeish, Alexandra Junza, Vanessa Soto-Cerrato, Petra Hyroššová, Miquel Angel Pujana, August Vidal, Agnès Figueras, Xavier Matias-Guiu, Sandra Orsulic, Jose C. Perales, Conxi Lázaro, Barbie Taylor-Harding, Diana Garzón, Pilar Barretina, Francesc Viñals, Joan Brunet, Cristina Muñoz-Pinedo, Oscar Yanes, Álvaro Lahiguera, Hisashi Tanaka, Violeta Serra, Javier A. Menendez, Luis Palomero, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, Institut Català de la Salut, [Lahiguera Á, Figueras A, Garzón D, Moreno R] Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain. Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain. [Hyroššová P] Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain. [Soto-Cerrato V] Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain. [Serra V] Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. CIBERONC, Instituto de Salud Carlos III, Madrid, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,Genome instability ,Medicine (General) ,Cancer cells ,Medicaments antineoplàstics - Ús terapèutic ,BCRA ,cancer metabolism ,neoplasias::neoplasias por localización::neoplasias de las glándulas endocrinas::neoplasias ováricas [ENFERMEDADES] ,QH426-470 ,Carcinoma, Ovarian Epithelial ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Oxidació ,Medical and Health Sciences ,0302 clinical medicine ,Ovarian Epithelial ,2.1 Biological and endogenous factors ,Aetiology ,Homologous Recombination ,Càncer ,PARP inhibitors ,11 Medical and Health Sciences ,Cancer ,Ovarian Neoplasms ,Chemistry ,Enzyme inhibitors ,Articles ,Biological Sciences ,OXPHOS ,Metabolisme ,Ovaris - Càncer ,5.1 Pharmaceuticals ,Molecular Medicine ,Female ,Cèl·lules canceroses ,Development of treatments and therapeutic interventions ,Biotechnology ,DNA repair ,Poly ADP ribose polymerase ,Genetic Phenomena::Recombination, Genetic::Homologous Recombination [PHENOMENA AND PROCESSES] ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Oxidative phosphorylation ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,03 medical and health sciences ,R5-920 ,Oxidation ,Genetics ,medicine ,Humans ,Recombinació genètica ,fenómenos genéticos::recombinación genética::recombinación homóloga [FENÓMENOS Y PROCESOS] ,Tumors ,Neoplasms::Neoplasms by Site::Endocrine Gland Neoplasms::Ovarian Neoplasms [DISEASES] ,Carcinoma ,06 Biological Sciences ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,Metabolism ,Inhibidors enzimàtics ,Cancer cell ,Cancer research ,NAD+ kinase ,metformin ,Homologous recombination ,030217 neurology & neurosurgery - Abstract
Mitochondrial metabolism and the generation of reactive oxygen species (ROS) contribute to the acquisition of DNA mutations and genomic instability in cancer. How genomic instability influences the metabolic capacity of cancer cells is nevertheless poorly understood. Here, we show that homologous recombination‐defective (HRD) cancers rely on oxidative metabolism to supply NAD + and ATP for poly(ADP‐ribose) polymerase (PARP)‐dependent DNA repair mechanisms. Studies in breast and ovarian cancer HRD models depict a metabolic shift that includes enhanced expression of the oxidative phosphorylation (OXPHOS) pathway and its key components and a decline in the glycolytic Warburg phenotype. Hence, HRD cells are more sensitive to metformin and NAD + concentration changes. On the other hand, shifting from an OXPHOS to a highly glycolytic metabolism interferes with the sensitivity to PARP inhibitors (PARPi) in these HRD cells. This feature is associated with a weak response to PARP inhibition in patient‐derived xenografts, emerging as a new mechanism to determine PARPi sensitivity. This study shows a mechanistic link between two major cancer hallmarks, which in turn suggests novel possibilities for specifically treating HRD cancers with OXPHOS inhibitors., Homologous recombination‐defective (HRD) cancers need high levels of NAD + and ATP for alternative PARP‐dependent DNA repair. HRD cancer cells undergo a characteristic metabolic shift that include enhanced OXPHOS, opening new opportunities for treatment with OXPHOS inhibitors like metformin.
- Published
- 2020
49. Generation of orthotopic pancreatic tumors and ex vivo characterization of tumor-infiltrating T cell cytotoxicity
- Author
-
Sarah, Spear, Iain A, McNeish, Melania, Capasso, and Ovarian Cancer Action
- Subjects
immunology [T-Lymphocytes, Cytotoxic] ,1702 Cognitive Sciences ,metabolism [Cytokines] ,Antineoplastic Agents ,metabolism [Antineoplastic Agents] ,Cell Separation ,Flow Cytometry ,0601 Biochemistry and Cell Biology ,Pancreatic Neoplasms ,metabolism [Pancreatic Neoplasms] ,Mice ,immunology [Pancreatic Neoplasms] ,1701 Psychology ,ddc:570 ,immunology [Tumor Microenvironment] ,Tumor Microenvironment ,Animals ,Humans ,Tetradecanoylphorbol Acetate ,Cytokines ,Neoplasm Transplantation ,T-Lymphocytes, Cytotoxic ,pathology [Pancreatic Neoplasms] - Abstract
In vivo models of pancreatic cancer provide invaluable tools for studying disease dynamics, immune infiltration and new therapeutic strategies. The orthotopic murine model can be performed on large cohorts of immunocompetent mice simultaneously, is relatively inexpensive and preserves the cognate tissue microenvironment. The quantification of T cell infiltration and cytotoxic activity within orthotopic tumors provides a useful indicator of an antitumoral response. This protocol describes the methodology for surgical generation of orthotopic pancreatic tumors by injection of a low number of syngeneic tumor cells resuspended in 5 µL basement membrane directly into the pancreas. Mice bearing orthotopic tumors take approximately 30 days to reach endpoint, at which point tumors can be harvested and processed for characterization of tumor-infiltrating T cell activity. Rapid enzymatic digestion using collagenase and DNase allows a single-cell suspension to be extracted from tumors. The viability and cell surface markers of immune cells extracted from the tumor are preserved; therefore, it is appropriate for multiple downstream applications, including flow-assisted cell sorting of immune cells for culture or RNA extraction, flow cytometry analysis of immune cell populations. Here, we describe the ex vivo stimulation of T cell populations for intracellular cytokine quantification (IFNγ and TNFα) and degranulation activity (CD107a) as a measure of overall cytotoxicity. Whole-tumor digests were stimulated with phorbol myristate acetate and ionomycin for 5 h, in the presence of anti-CD107a antibody in order to upregulate cytokine production and degranulation. The addition of brefeldin A and monensin for the final 4 h was performed to block extracellular transport and maximize cytokine detection. Extra- and intra-cellular staining of cells was then performed for flow cytometry analysis, where the proportion of IFNγ+, TNFα+ and CD107a+ CD4+ and CD8+ T cells was quantified. This method provides a starting base to perform comprehensive analysis of the tumor microenvironment.
- Published
- 2019
50. Safety and efficacy of the tumor-selective adenovirus enadenotucirev with or without paclitaxel in platinum-resistant ovarian cancer: a phase 1 clinical trial
- Author
-
Moreno, Victor, Barretina-Ginesta, Maria-Pilar, García-Donas, Jesús, Jayson, Gordon C, Roxburgh, Patricia, Vázquez, Raúl Márquez, Michael, Agnieszka, Antón-Torres, Antonio, Brown, Richard, Krige, David, Champion, Brian, McNeish, Iain, Imperial College Healthcare NHS Trust- BRC Funding, Ovarian Cancer Action, Cancer Research UK, and National Institute for Health Research
- Subjects
Adult ,Male ,Cancer Research ,Maximum Tolerated Dose ,Paclitaxel ,Immunology ,Drug Evaluation, Preclinical ,therapies ,Carcinoma, Ovarian Epithelial ,investigational ,Adenoviridae ,Mice ,Animals ,Humans ,Immunology and Allergy ,Aged ,Platinum ,Ovarian Neoplasms ,Pharmacology ,clinical trials as topic ,Middle Aged ,Prognosis ,Antineoplastic Agents, Phytogenic ,Combined Modality Therapy ,Survival Rate ,Oncolytic and Local Immunotherapy ,Oncology ,Drug Resistance, Neoplasm ,oncolytic viruses ,Molecular Medicine ,Female ,immunotherapy ,Follow-Up Studies - Abstract
BackgroundTreatment outcomes remain poor in recurrent platinum-resistant ovarian cancer. Enadenotucirev, a tumor-selective and blood stable adenoviral vector, has demonstrated a manageable safety profile in phase 1 studies in epithelial solid tumors.MethodsWe conducted a multicenter, open-label, phase 1 dose-escalation and dose-expansion study (OCTAVE) to assess enadenotucirev plus paclitaxel in patients with platinum-resistant epithelial ovarian cancer. During phase 1a, the maximum tolerated dose of intraperitoneally administered enadenotucirev monotherapy (three doses; days 1, 8 and 15) was assessed using a 3+3 dose-escalation model. Phase 1b included a dose-escalation and an intravenous dosing dose-expansion phase assessing enadenotucirev plus paclitaxel. For phase 1a/b, the primary objective was to determine the maximum tolerated dose of enadenotucirev (with paclitaxel in phase 1b). In the dose-expansion phase, the primary endpoint was progression-free survival (PFS). Additional endpoints included response rate and T-cell infiltration.ResultsOverall, 38 heavily pretreated patients were enrolled and treated. No dose-limiting toxicities were observed at any doses. However, frequent catheter complications led to the discontinuation of intraperitoneal dosing during phase 1b. Intravenous enadenotucirev (1×1012 viral particles; days 1, 3 and 5 every 28-days for two cycles) plus paclitaxel (80 mg/m2; days 9, 16 and 23 of each cycle) was thus selected for dose-expansion. Overall, 24/38 (63%) patients experienced at least 1 Grade ≥3 treatment-emergent adverse event (TEAE); most frequently neutropenia (21%). Six patients discontinued treatment due to TEAEs, including one patient due to a grade 2 treatment-emergent serious AE of catheter site infection (intraperitoneal enadenotucirev monotherapy). Among the 20 patients who received intravenous enadenotucirev plus paclitaxel, 4-month PFS rate was 64% (median 6.2 months), objective response rate was 10%, 35% of patients achieved stable disease and 65% of patients had a reduction in target lesion burden at ≥1 time point. Five out of six patients with matched pre-treatment and post-treatment biopsies treated with intravenous enadenotucirev plus paclitaxel had increased (mean 3.1-fold) infiltration of CD8 +T cells in post-treatment biopsies.ConclusionsIntravenously dosed enadenotucirev plus paclitaxel demonstrated manageable tolerability, an encouraging median PFS and increased tumor immune-cell infiltration in platinum-resistant ovarian cancer.Trial registration numberNCT02028117.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.