20 results on '"Toby Prout"'
Search Results
2. A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Author
-
Susana Banerjee, Vasiliki Michalarea, Joo Ern Ang, Alvaro Ingles Garces, Andrea Biondo, Ionut-Gabriel Funingana, Martin Little, Ruth Ruddle, Florence Raynaud, Ruth Riisnaes, Bora Gurel, Sue Chua, Nina Tunariu, Joanna C. Porter, Toby Prout, Mona Parmar, Anna Zachariou, Alison Turner, Ben Jenkins, Stuart McIntosh, Ed Ainscow, Anna Minchom, Juanita Lopez, Johann de Bono, Robert Jones, Emma Hall, Natalie Cook, Bristi Basu, Udai Banerji, Banerjee, Susana [0000-0002-8840-7934], Michalarea, Vasiliki [0000-0002-3102-3534], Ang, Joo Ern [0000-0003-2103-996X], Ingles Garces, Alvaro [0000-0002-0073-4237], Biondo, Andrea [0000-0003-0599-254X], Funingana, Ionut-Gabriel [0000-0002-1197-2652], Little, Martin [0000-0003-2592-1570], Ruddle, Ruth [0000-0003-0025-8872], Raynaud, Florence [0000-0003-0957-6279], Riisnaes, Ruth [0000-0002-8924-302X], Gurel, Bora [0000-0002-5018-8078], Chua, Sue [0000-0001-5369-8156], Tunariu, Nina [0000-0001-6656-3699], Porter, Joanna C [0000-0002-7307-169X], Prout, Toby [0000-0002-6465-4578], Parmar, Mona [0000-0001-7818-4100], Zachariou, Anna [0000-0002-7867-8327], Turner, Alison [0000-0003-2915-2756], Jenkins, Ben [0000-0002-2517-3595], McIntosh, Stuart [0000-0002-7379-4505], Ainscow, Ed [0000-0002-3119-8422], Minchom, Anna [0000-0002-9339-7101], Lopez, Juanita [0000-0001-8321-4212], de Bono, Johann [0000-0002-2034-595X], Jones, Robert [0000-0003-3576-9496], Hall, Emma [0000-0001-5999-5020], Cook, Natalie [0000-0003-2606-1082], Basu, Bristi [0000-0002-3562-2868], Banerji, Udai [0000-0003-1503-3123], and Apollo - University of Cambridge Repository
- Subjects
Ovarian Neoplasms ,Cancer Research ,Folic Acid ,Maximum Tolerated Dose ,Oncology ,Neoplasms ,Humans ,Female ,Thymidylate Synthase ,Enzyme Inhibitors - Abstract
Purpose: CT900 is a novel small molecule thymidylate synthase inhibitor that binds to α-folate receptor (α-FR) and thus is selectively taken up by α-FR–overexpressing tumors. Patients and Methods: A 3+3 dose escalation design was used. During dose escalation, CT900 doses of 1–6 mg/m2 weekly and 2–12 mg/m2 every 2 weeks (q2Wk) intravenously were evaluated. Patients with high-grade serous ovarian cancer were enrolled in the expansion cohorts. Results: 109 patients were enrolled: 42 patients in the dose escalation and 67 patients in the expansion cohorts. At the dose/schedule of 12 mg/m2/q2Wk (with and without dexamethasone, n = 40), the most common treatment-related adverse events were fatigue, nausea, diarrhea, cough, anemia, and pneumonitis, which were predominantly grade 1 and grade 2. Levels of CT900 more than 600 nmol/L needed for growth inhibition in preclinical models were achieved for >65 hours at a dose of 12 mg/m2. In the expansion cohorts, the overall response rate (ORR), was 14/64 (21.9%). Thirty-eight response-evaluable patients in the expansion cohorts receiving 12 mg/m2/q2Wk had tumor evaluable for quantification of α-FR. Patients with high or medium expression had an objective response rate of 9/25 (36%) compared with 1/13 (7.7%) in patients with negative/very low or low expression of α-FR. Conclusions: The dose of 12 mg/m2/q2Wk was declared the recommended phase II dose/schedule. At this dose/schedule, CT900 exhibited an acceptable side effect profile with clinical benefit in patients with high/medium α-FR expression and warrants further investigation.
- Published
- 2022
3. Figure S4 from Triplet Therapy with Palbociclib, Taselisib, and Fulvestrant in PIK3CA-Mutant Breast Cancer and Doublet Palbociclib and Taselisib in Pathway-Mutant Solid Cancers
- Author
-
Nicholas C. Turner, Juanita S. Lopez, Timothy A. Yap, Emma Hall, Alison Turner, Florence I. Raynaud, Ruth Ruddle, Karen E. Swales, Jason Malia, Laura Finneran, Toby Prout, Mona Parmar, Jenny King, Margaret Hills, Michael Hubank, Paula Z. Proszek, Heidrun Gevensleben, Sarah Hrebien, Alex Pearson, Isaac Garcia-Murillas, Maria Teresa Herrera-Abreu, Rosalind J. Cutts, Alicia F.C. Okines, Alistair Ring, Anne C. Armstrong, Iain R. Macpherson, Joline S.J. Lim, and Javier Pascual
- Abstract
Baseline tissue biomarker analysis
- Published
- 2023
4. Table S1 from Triplet Therapy with Palbociclib, Taselisib, and Fulvestrant in PIK3CA-Mutant Breast Cancer and Doublet Palbociclib and Taselisib in Pathway-Mutant Solid Cancers
- Author
-
Nicholas C. Turner, Juanita S. Lopez, Timothy A. Yap, Emma Hall, Alison Turner, Florence I. Raynaud, Ruth Ruddle, Karen E. Swales, Jason Malia, Laura Finneran, Toby Prout, Mona Parmar, Jenny King, Margaret Hills, Michael Hubank, Paula Z. Proszek, Heidrun Gevensleben, Sarah Hrebien, Alex Pearson, Isaac Garcia-Murillas, Maria Teresa Herrera-Abreu, Rosalind J. Cutts, Alicia F.C. Okines, Alistair Ring, Anne C. Armstrong, Iain R. Macpherson, Joline S.J. Lim, and Javier Pascual
- Abstract
RMH200 panel design
- Published
- 2023
5. Data from Triplet Therapy with Palbociclib, Taselisib, and Fulvestrant in PIK3CA-Mutant Breast Cancer and Doublet Palbociclib and Taselisib in Pathway-Mutant Solid Cancers
- Author
-
Nicholas C. Turner, Juanita S. Lopez, Timothy A. Yap, Emma Hall, Alison Turner, Florence I. Raynaud, Ruth Ruddle, Karen E. Swales, Jason Malia, Laura Finneran, Toby Prout, Mona Parmar, Jenny King, Margaret Hills, Michael Hubank, Paula Z. Proszek, Heidrun Gevensleben, Sarah Hrebien, Alex Pearson, Isaac Garcia-Murillas, Maria Teresa Herrera-Abreu, Rosalind J. Cutts, Alicia F.C. Okines, Alistair Ring, Anne C. Armstrong, Iain R. Macpherson, Joline S.J. Lim, and Javier Pascual
- Abstract
Cyclin-dependent kinase 4/6 (CDK4/6) and PI3K inhibitors synergize in PIK3CA-mutant ER-positive HER2-negative breast cancer models. We conducted a phase Ib trial investigating the safety and efficacy of doublet CDK4/6 inhibitor palbociclib plus selective PI3K inhibitor taselisib in advanced solid tumors, and triplet palbociclib plus taselisib plus fulvestrant in 25 patients with PIK3CA-mutant, ER-positive HER2-negative advanced breast cancer. The triplet therapy response rate in PIK3CA-mutant, ER-positive HER2-negative cancer was 37.5% [95% confidence interval (CI), 18.8–59.4]. Durable disease control was observed in PIK3CA-mutant ER-negative breast cancer and other solid tumors with doublet therapy. Both combinations were well tolerated at pharmacodynamically active doses. In the triplet group, high baseline cyclin E1 expression associated with shorter progression-free survival (PFS; HR = 4.2; 95% CI, 1.3–13.1; P = 0.02). Early circulating tumor DNA (ctDNA) dynamics demonstrated high on-treatment ctDNA association with shorter PFS (HR = 5.2; 95% CI, 1.4–19.4; P = 0.04). Longitudinal plasma ctDNA sequencing provided genomic evolution evidence during triplet therapy.SIGNIFICANCE:The triplet of palbociclib, taselisib, and fulvestrant has promising efficacy in patients with heavily pretreated PIK3CA-mutant ER-positive HER2-negative advanced breast cancer. A subset of patients with PIK3CA-mutant triple-negative breast cancer derived clinical benefit from palbociclib and taselisib doublet, suggesting a potential nonchemotherapy targeted approach for this population.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2023
6. Supplementary Figure from A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Author
-
Udai Banerji, Bristi Basu, Natalie Cook, Emma Hall, Robert Jones, Johann de Bono, Juanita Lopez, Anna Minchom, Ed Ainscow, Stuart McIntosh, Ben Jenkins, Alison Turner, Anna Zachariou, Mona Parmar, Toby Prout, Joanna C. Porter, Nina Tunariu, Sue Chua, Bora Gurel, Ruth Riisnaes, Florence Raynaud, Ruth Ruddle, Martin Little, Ionut-Gabriel Funingana, Andrea Biondo, Alvaro Ingles Garces, Joo Ern Ang, Vasiliki Michalarea, and Susana Banerjee
- Abstract
Supplementary Figure from A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Published
- 2023
7. Supplementary Table from A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Author
-
Udai Banerji, Bristi Basu, Natalie Cook, Emma Hall, Robert Jones, Johann de Bono, Juanita Lopez, Anna Minchom, Ed Ainscow, Stuart McIntosh, Ben Jenkins, Alison Turner, Anna Zachariou, Mona Parmar, Toby Prout, Joanna C. Porter, Nina Tunariu, Sue Chua, Bora Gurel, Ruth Riisnaes, Florence Raynaud, Ruth Ruddle, Martin Little, Ionut-Gabriel Funingana, Andrea Biondo, Alvaro Ingles Garces, Joo Ern Ang, Vasiliki Michalarea, and Susana Banerjee
- Abstract
Supplementary Table from A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Published
- 2023
8. Data from A Phase I Trial of CT900, a Novel α-Folate Receptor–Mediated Thymidylate Synthase Inhibitor, in Patients with Solid Tumors with Expansion Cohorts in Patients with High-Grade Serous Ovarian Cancer
- Author
-
Udai Banerji, Bristi Basu, Natalie Cook, Emma Hall, Robert Jones, Johann de Bono, Juanita Lopez, Anna Minchom, Ed Ainscow, Stuart McIntosh, Ben Jenkins, Alison Turner, Anna Zachariou, Mona Parmar, Toby Prout, Joanna C. Porter, Nina Tunariu, Sue Chua, Bora Gurel, Ruth Riisnaes, Florence Raynaud, Ruth Ruddle, Martin Little, Ionut-Gabriel Funingana, Andrea Biondo, Alvaro Ingles Garces, Joo Ern Ang, Vasiliki Michalarea, and Susana Banerjee
- Abstract
Purpose:CT900 is a novel small molecule thymidylate synthase inhibitor that binds to α-folate receptor (α-FR) and thus is selectively taken up by α-FR–overexpressing tumors.Patients and Methods:A 3+3 dose escalation design was used. During dose escalation, CT900 doses of 1–6 mg/m2 weekly and 2–12 mg/m2 every 2 weeks (q2Wk) intravenously were evaluated. Patients with high-grade serous ovarian cancer were enrolled in the expansion cohorts.Results:109 patients were enrolled: 42 patients in the dose escalation and 67 patients in the expansion cohorts. At the dose/schedule of 12 mg/m2/q2Wk (with and without dexamethasone, n = 40), the most common treatment-related adverse events were fatigue, nausea, diarrhea, cough, anemia, and pneumonitis, which were predominantly grade 1 and grade 2. Levels of CT900 more than 600 nmol/L needed for growth inhibition in preclinical models were achieved for >65 hours at a dose of 12 mg/m2. In the expansion cohorts, the overall response rate (ORR), was 14/64 (21.9%). Thirty-eight response-evaluable patients in the expansion cohorts receiving 12 mg/m2/q2Wk had tumor evaluable for quantification of α-FR. Patients with high or medium expression had an objective response rate of 9/25 (36%) compared with 1/13 (7.7%) in patients with negative/very low or low expression of α-FR.Conclusions:The dose of 12 mg/m2/q2Wk was declared the recommended phase II dose/schedule. At this dose/schedule, CT900 exhibited an acceptable side effect profile with clinical benefit in patients with high/medium α-FR expression and warrants further investigation.
- Published
- 2023
9. Abstract CT093: Preliminary evidence of antitumor activity of Ipatasertib (Ipat) and Atezolizumab (A) in glioblastoma (GBM) patients (pts) with PTEN loss in the Phase 1 Ice-CAP trial (NCT03673787)
- Author
-
Crescens Tiu, Wing Hing Yau, Liam C. Welsh, Timothy L. Jones, Anna Zachariou, Toby Prout, Mona Parmar, Alison J. Turner, Robert W. Daly, Christina Yap, Mateus Crespo, Bora Gurel, Ruth M. Riisnaes, Shaun A. DeCordova, Karen E. Swales, Udai Banerji, Anna R. Minchom, Adam Sharp, Johann S. de Bono, and Juanita S. Lopez
- Subjects
Cancer Research ,Oncology - Abstract
Background PTEN loss of function is frequent in GBM correlating with poor prognosis, impaired antitumor responses and reduced efficacy of Immune Checkpoint Inhibitors (ICI). Ipat is a potent, selective, small-molecule inhibitor of Akt. Ipat efficiently depletes FOXP3+ regulatory T cells from the tumor microenvironment (TME) resulting in increased infiltration of effector T cells in solid tumors (Lopez 2020, AACR). We hypothesize that Akt inhibition in PTEN loss glioblastomas may deplete the TME of suppressive immune cells, and render malignant brain tumors more responsive to ICIs. Methods Relapsed WHO grade IV GBM pts with stable neurological symptoms ≥5 days prior to enrollment, requiring Results 23 recurrent GBM pts (median age 55 yrs (25-71 yrs; 74% male) were enrolled. Median ECOG PS 1. All pts had surgery followed by radical-chemoradiotherapy. Median prior lines of therapy 1 (range 1-4). 15 pts had PTEN loss by IHC (H 6 cycles) in 19 efficacy evaluable pts was 32% (6/19 overall) and 28.6% (4/14) for pts with PTEN loss. Multiplex IHC of archival samples (n=19) showed PTEN loss tumors had significantly greater numbers of CD3+ infiltrating T-lymphocytes within the TME compared to PTEN wild type tumors (median 53.64/mm2 vs 1.94/mm2; p=0.0021). Within the PTEN loss cohort (n=13), pts with clinical benefit had significantly higher baseline number of CD8+ effector T cells as compared to non-responders (median 46.50/mm2 vs 8.21/mm2; p=0.0336). Two pts with PTEN loss proceeded to re-resection on trial. One exceptional responder, a 58yr male refractory to radical chemo-radiotherapy and Bevacizumab had a resection of enhancing disease after 5 cycles that showed >70% depletion of CD4+ T regulatory cells with an increase in CD8+ lymphocyte infiltration and no residual evidence of tumor (pathological CR). In contrast, a non-responding patient who proceeded to debulking surgery had no change in infiltration of CD8+ lymphocytes, but a marked increase (>27-fold) in CD4+ regulatory T cells. Conclusion The RP2D of 400mg Ipa OD + 1200mg A Q3W was well tolerated in GBM pts. Early efficacy signals were detected with PTEN loss being a promising predictive biomarker for response to combination. Citation Format: Crescens Tiu, Wing Hing Yau, Liam C. Welsh, Timothy L. Jones, Anna Zachariou, Toby Prout, Mona Parmar, Alison J. Turner, Robert W. Daly, Christina Yap, Mateus Crespo, Bora Gurel, Ruth M. Riisnaes, Shaun A. DeCordova, Karen E. Swales, Udai Banerji, Anna R. Minchom, Adam Sharp, Johann S. de Bono, Juanita S. Lopez. Preliminary evidence of antitumor activity of Ipatasertib (Ipat) and Atezolizumab (A) in glioblastoma (GBM) patients (pts) with PTEN loss in the Phase 1 Ice-CAP trial (NCT03673787) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT093.
- Published
- 2023
10. Abstract 3461: Preclinical and clinical evaluation of the RAF/MEK clamp avutometinib (VS-6766) in combination with the mTOR inhibitor everolimus for the treatment of KRAS mutated non-small cell lung cancer
- Author
-
Simon Rodney, Adam R. Stewart, Victoria Sanchez Perez, Cienne Morton, Lisa A. Pickard, Taleen Shakouri, Toby Prout, Mona Parmar, Alison J. Turner, Silvia Coma, Jonathan Pachter, Laura Finneran, Emma Hall, James Spicer, Anna Minchom, and Udai Banerji
- Subjects
Cancer Research ,Oncology - Abstract
Background: KRAS mutations (mt) are found in ~30% of non-small cell lung cancers (NSCLC). Despite the approval and development of KRAS G12C and KRAS G12D inhibitors respectively, mechanisms of resistance to MAPK pathway inhibitors are emerging and combination strategies are needed for patients with KRAS mt NSCLC . Avutometinib (VS-6766) is a unique RAF/MEK clamp that inhibits MEK kinase activity and blocks RAF-mediated phosphorylation of MEK. We studied the preclinical and clinical activity of the combination of avutometinib with the mTOR inhibitor everolimus in KRAS mt NSCLC. Methods: A panel of KRAS mt NSCLC cell lines were treated for 1 hour with clinically relevant concentrations of avutometinib and everolimus and changes in phosphoproteins were measured using an antibody array. We then tested for synergy of avutometinib and everolimus in 3D proliferation assays and in the H441 NSCLC xenograft model. A clinical trial is ongoing (NCT02407509) testing the combination of 3.2 mg avutometinib with 5 mg of everolimus administered twice weekly 3 weeks on/1 week off in 28-day cycles (previously defined as the recommended phase 2 dose) in a cohort of patients with KRAS mt NSCLC. Results: Avutometinib inhibited the MAPK pathway (p-MEK, p-ERK, p-90RSK) with an increase in p-PRAS40, suggesting activation of the PI3K pathway as an adaptive resistance mechanism. Everolimus inhibited the PI3K pathway (p-p70S6K and p-RPS6). Among a panel of KRAS mt NSCLC cell lines, avutometinib + everolimus showed synergistic anti-proliferative activity across KRAS G12C, G12V and G12D variants (mean synergy score of ~18). In the H441 KRAS G12V NSCLC xenograft model, there was a significant reduction in tumor volume and increase in survival with the combination of avutometinib and everolimus (87% TGI; 66 days vs 36.5 days in control group) compared to control. In the clinical trial expansion, 16 patients with KRAS mt NSCLC have been treated so far with avutometinib and everolimus (5 G12V, 3 Q61H, 2 G12C, 2 G12A, 2 G12D, 1 G13A, 1 G13D; median prior lines = 2). The current objective response rate (ORR) among the 14 patients who have at least one post assessment scan is 3/14 (21%; 1 G12V, 1 G12A, 1 G13A) with 11/14 showing a reduction in tumor size as best response. The current progression free survival (PFS) is 5.3 months (95% CI 2.8-7.4 months) with 4 patients still on study. Updated data on the planned cohort size of 20 patients will be presented. Conclusion: The combination of avutometinib and everolimus overcomes the activation of the PI3K/AKT/mTOR pathway which is an adaptive resistance mechanism to MAPK pathway inhibition. We have shown that avutometinib and everolimus induce synergistic anti-tumor effects preclinically, and preliminary data suggest clinically meaningful ORR and PFS in patients with KRAS mt NSCLC including non-G12C variants. Citation Format: Simon Rodney, Adam R. Stewart, Victoria Sanchez Perez, Cienne Morton, Lisa A. Pickard, Taleen Shakouri, Toby Prout, Mona Parmar, Alison J. Turner, Silvia Coma, Jonathan Pachter, Laura Finneran, Emma Hall, James Spicer, Anna Minchom, Udai Banerji. Preclinical and clinical evaluation of the RAF/MEK clamp avutometinib (VS-6766) in combination with the mTOR inhibitor everolimus for the treatment of KRAS mutated non-small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3461.
- Published
- 2023
11. Triplet Therapy with Palbociclib, Taselisib, and Fulvestrant inPIK3CA-Mutant Breast Cancer and Doublet Palbociclib and Taselisib in Pathway-Mutant Solid Cancers
- Author
-
Heidrun Gevensleben, Alistair Ring, Toby Prout, Nicholas C. Turner, Alison Turner, Iain R. Macpherson, Paula Proszek, Alex Pearson, Mike Hubank, Rosalind J. Cutts, Karen E Swales, Sarah Hrebien, Javier Pascual, Margaret Hills, Maria Teresa Herrera-Abreu, Jenny King, Jason Malia, Anne C Armstrong, Ruth Ruddle, Alicia Okines, Isaac Garcia-Murillas, Emma Hall, Mona Parmar, Laura Finneran, Florence I. Raynaud, Timothy A. Yap, Juanita Lopez, and Joline S.J. Lim
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,education.field_of_study ,Fulvestrant ,Kinase ,business.industry ,Population ,Cancer ,Palbociclib ,medicine.disease ,03 medical and health sciences ,Cyclin E1 ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,education ,business ,neoplasms ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Cyclin-dependent kinase 4/6 (CDK4/6) and PI3K inhibitors synergize in PIK3CA-mutant ER-positive HER2-negative breast cancer models. We conducted a phase Ib trial investigating the safety and efficacy of doublet CDK4/6 inhibitor palbociclib plus selective PI3K inhibitor taselisib in advanced solid tumors, and triplet palbociclib plus taselisib plus fulvestrant in 25 patients with PIK3CA-mutant, ER-positive HER2-negative advanced breast cancer. The triplet therapy response rate in PIK3CA-mutant, ER-positive HER2-negative cancer was 37.5% [95% confidence interval (CI), 18.8–59.4]. Durable disease control was observed in PIK3CA-mutant ER-negative breast cancer and other solid tumors with doublet therapy. Both combinations were well tolerated at pharmacodynamically active doses. In the triplet group, high baseline cyclin E1 expression associated with shorter progression-free survival (PFS; HR = 4.2; 95% CI, 1.3–13.1; P = 0.02). Early circulating tumor DNA (ctDNA) dynamics demonstrated high on-treatment ctDNA association with shorter PFS (HR = 5.2; 95% CI, 1.4–19.4; P = 0.04). Longitudinal plasma ctDNA sequencing provided genomic evolution evidence during triplet therapy.SIGNIFICANCE:The triplet of palbociclib, taselisib, and fulvestrant has promising efficacy in patients with heavily pretreated PIK3CA-mutant ER-positive HER2-negative advanced breast cancer. A subset of patients with PIK3CA-mutant triple-negative breast cancer derived clinical benefit from palbociclib and taselisib doublet, suggesting a potential nonchemotherapy targeted approach for this population.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2021
12. Abstract 3476: Mechanistic evaluation of VS-6766 (dual RAF/MEK inhibitor) and defactinib (FAK inhibitor) in low-grade serous ovarian cancer models with correlations to clinical response
- Author
-
Adam R. Stewart, Lisa Pickard, Ekta Paranjape, Victoria Sanchez Perez, Sanjib Chowdhury, Stephanie Lustgarten, Silvia Coma, Jonathan A. Pachter, Mark S. Carey, Gabriel DiMattia, Hannah C. Badham, Toby Prout, Mona Parmar, Muneeb Mahmud, Christina Yap, Matthew G. Krebs, Susana Banerjee, and Udai Banerji
- Subjects
Cancer Research ,Oncology - Abstract
Background: Low-grade serous ovarian cancer (LGSOC) constitutes up to 10% of all ovarian cancer and has clinical and molecular characteristics (resistance to chemotherapy, presence of RAS/RAF mutations, lack of TP53 mutations) distinct from high-grade serous ovarian cancer. Here, we characterized the effects of the dual RAF/MEK inhibitor VS-6766 and the FAK inhibitor defactinib on signal transduction and viability in LGSOC cell lines and patient-derived organoids. To correlate molecular characteristics with clinical response, we characterized genomic alterations in archival tumor samples from patients with LGSOC treated with the combination of VS-6766 and defactinib on a clinical trial (FRAME). Material and Methods: We exposed 5 LGSOC cell lines to clinical Cmax concentrations adjusted for protein binding of VS-6766 and defactinib. We quantified phospho- and total proteins (n=66) with an antibody-bead based assay normalized to GAPDH. We also studied growth inhibitory effects of the combination on KRAS mutant (mt) LGSOC patient-derived organoids. We performed next generation sequencing on archival samples from LGSOC patients treated with VS-6766 in combination with defactinib. Results: Signal transduction changes at 1 hr included reduction of p-FAK in 5/5 cell lines in response to defactinib. Cells exposed to VS-6766 showed a reduction in p-ERK and p-p90-RSK in 4/5 cell lines. Additionally, VS-6766 decreased p-cJUN and increased p-IκB in 4/5 cell lines, changes correlated with apoptosis. At 24 hrs, p-ERK and p-p90-RSK inhibition were maintained in 3/5 cell lines. Both drugs increased cleaved PARP in 4/5 cell lines and VS-6766 increased p-SMAD3 and BIM levels, indicating an increase in cell death/apoptosis. The combination of VS-6766 + defactinib showed synergistic growth inhibition in a KRAS mt LGSOC organoid model (combination index 0.51). The clinical combination of VS-6766 and defactinib (September 2021 cut-off) has shown an objective response rate (ORR) of 11/24 (46%) across all patients with LGSOC, and an ORR of 64% (7/11) for patients with KRAS mt LGSOC (n=11). In addition to mutations in KRAS, emerging data may suggest a correlation of U2AF1 and MED12 mutations with response. Conclusions: VS-6766, the dual RAF/MEK inhibitor, induces significant inhibition of ERK pathway signaling in addition to perturbations in TNF/NFκB signaling. Both defactinib and VS-6766 induce apoptosis in LGSOC models. The results provide mechanistic insights into the encouraging response rates observed in patients with LGSOC treated with VS-6766 and defactinib (NCT03875820). These data support the ongoing randomized phase II ENGOTov60/GOG3052/RAMP201 study (NCT04625270). Citation Format: Adam R. Stewart, Lisa Pickard, Ekta Paranjape, Victoria Sanchez Perez, Sanjib Chowdhury, Stephanie Lustgarten, Silvia Coma, Jonathan A. Pachter, Mark S. Carey, Gabriel DiMattia, Hannah C. Badham, Toby Prout, Mona Parmar, Muneeb Mahmud, Christina Yap, Matthew G. Krebs, Susana Banerjee, Udai Banerji. Mechanistic evaluation of VS-6766 (dual RAF/MEK inhibitor) and defactinib (FAK inhibitor) in low-grade serous ovarian cancer models with correlations to clinical response [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3476.
- Published
- 2022
13. Phase 1, dose-escalation study of guadecitabine (SGI-110) in combination with pembrolizumab in patients with solid tumors
- Author
-
Dionysis Papadatos-Pastos, Wei Yuan, Abhijit Pal, Mateus Crespo, Ana Ferreira, Bora Gurel, Toby Prout, Malaka Ameratunga, Maxime Chénard-Poirier, Andra Curcean, Claudia Bertan, Chloe Baker, Susana Miranda, Nahal Masrour, Wentin Chen, Rita Pereira, Ines Figueiredo, Ricardo Morilla, Ben Jenkins, Anna Zachariou, Ruth Riisnaes, Mona Parmar, Alison Turner, Suzanne Carreira, Christina Yap, Robert Brown, Nina Tunariu, Udai Banerji, Juanita Lopez, Johann de Bono, and Anna Minchom
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Immunology ,Azacitidine ,Humans ,Molecular Medicine ,Immunology and Allergy ,Antibodies, Monoclonal, Humanized ,Immune Checkpoint Inhibitors - Abstract
BackgroundData suggest that immunomodulation induced by DNA hypomethylating agents can sensitize tumors to immune checkpoint inhibitors. We conducted a phase 1 dose-escalation trial (NCT02998567) of guadecitabine and pembrolizumab in patients with advanced solid tumors. We hypothesized that guadecitabine will overcome pembrolizumab resistance.MethodsPatients received guadecitabine (45 mg/m2 or 30 mg/m2, administered subcutaneously on days 1–4), with pembrolizumab (200 mg administered intravenously starting from cycle 2 onwards) every 3 weeks. Primary endpoints were safety, tolerability and maximum tolerated dose; secondary and exploratory endpoints included objective response rate (ORR), changes in methylome, transcriptome, immune contextures in pre-treatment and on-treatment tumor biopsies.ResultsBetween January 2017 and January 2020, 34 patients were enrolled. The recommended phase II dose was guadecitabine 30 mg/m2, days 1–4, and pembrolizumab 200 mg on day 1 every 3 weeks. Two dose-limiting toxicities (neutropenia, febrile neutropenia) were reported at guadecitabine 45 mg/m2 with none reported at guadecitabine 30 mg/m2. The most common treatment-related adverse events (TRAEs) were neutropenia (58.8%), fatigue (17.6%), febrile neutropenia (11.8%) and nausea (11.8%). Common, grade 3+ TRAEs were neutropaenia (38.2%) and febrile neutropaenia (11.8%). There were no treatment-related deaths. Overall, 30 patients were evaluable for antitumor activity; ORR was 7% with 37% achieving disease control (progression-free survival) for ≥24 weeks. Of 12 evaluable patients with non-small cell lung cancer, 10 had been previously treated with immune checkpoint inhibitors with 5 (42%) having disease control ≥24 weeks (clinical benefit). Reduction in LINE-1 DNA methylation following treatment in blood (peripheral blood mononuclear cells) and tissue samples was demonstrated and methylation at transcriptional start site and 5’ untranslated region gene regions showed enriched negative correlation with gene expression. Increases in intra-tumoural effector T-cells were seen in some responding patients. Patients having clinical benefit had high baseline inflammatory signature on RNAseq analyses.ConclusionsGuadecitabine in combination with pembrolizumab is tolerable with biological and anticancer activity. Reversal of previous resistance to immune checkpoint inhibitors is demonstrated.
- Published
- 2022
14. Phase I trial of the RAF/MEK clamp VS-6766 in combination with everolimus using an intermittent schedule with expansion in NSCLC across multiple KRAS variants
- Author
-
Anna Rachel Minchom, Vicky Sanchez Perez, Cienne Morton, Thubeena Manickavasagar, George Nintos, Julia Elizabeth Lai-Kwon, Christina Guo, Nina Tunariu, Tom Parker, Toby Prout, Mona Parmar, Alison Joanne Turner, Laura Finneran, Emma Hall, Jonathan A. Pachter, Louis J. Denis, James F. Spicer, and Udai Banerji
- Subjects
Cancer Research ,Oncology - Abstract
9018 Background: VS-6766 is a small molecule RAF/MEK clamp that results in the reduction of p-MEK and p-ERK. Preclinical data show synergy of VS-6766 with the mTOR inhibitor everolimus across a panel of KRAS mutated (mt) NSCLC cell lines. This clinical trial evaluated the safety and efficacy of a novel intermittent regimen of VS-6766 and everolimus with an expansion in KRAS mt NSCLC (NCT02407509). Methods: The trial used a 3+3 dose escalation design with an intermittent once a week schedule A, and if tolerated, twice a week schedule B (Mon-Thu or Tue-Fri) for both drugs on a 3 weeks on/1 week off, 28 day cycle. Patients with RAS or RAF mt cancers were eligible for the dose escalation cohort, and 20 patients with KRAS mt NSCLC will be treated in the dose expansion cohort. Toxicity was evaluated by NCI CTC V4 and efficacy was evaluated using RECIST 1.1. Results: A total of 28 patients have been treated; median age 60 yrs (range 36-78), and median lines of previous treatment 3 (range 0-7). Sixteen patients have been treated in the dose escalation (3 in schedule A and 13 in the schedule B). The doses of 4 mg of VS-6766 and 5 mg everolimus (once weekly) were tolerated with no dose limiting toxicities (DLTs) and the dose intensity escalated to schedule B (twice weekly). At 4 mg VS-6766 twice weekly, DLTs were observed in two out of six patients and included grade 4 CPK elevation and grade 3 rash. Thus, the dose in schedule B (twice weekly) was de-escalated to 3.2 mg VS-6766 and the dose of everolimus was kept at 5 mg. No DLTs were reported in 6 patients and thus this was declared as the recommended phase 2 dose (RP2D). At the RP2D, the grade 3-4 drug related AE were rash (18%) and pruritus (7%). In the dose escalation cohorts, 3 partial responses (PRs) were reported (2 KRAS G12D low grade serous ovarian cancer and 1 NRAS Q61K mt thyroid cancer). In the KRAS mt NSCLC expansion cohort, 10 patients are evaluable for efficacy and 2 confirmed responses were reported ( KRAS mutations G12V and G13A) with an objective response rate (ORR) 20% to date. The disease control rate (PR + SD) at the first scheduled evaluation was 90%. The median progression free survival (PFS) in the KRAS mt NSCLC cohort is 6.35 months (95% CI 3.52 – not reached). Updated ORR and PFS data will be presented. Conclusions: A tolerable intermittent dosing schedule targeting both the MAPK and PI3K pathways has been established. The combination of VS-6766 with everolimus has shown activity in patients with a variety of KRAS mutation variants including responses in KRAS mt NSCLC.
- Published
- 2022
15. Triplet Therapy with Palbociclib, Taselisib, and Fulvestrant in
- Author
-
Javier, Pascual, Joline S J, Lim, Iain R, Macpherson, Anne C, Armstrong, Alistair, Ring, Alicia F C, Okines, Rosalind J, Cutts, Maria Teresa, Herrera-Abreu, Isaac, Garcia-Murillas, Alex, Pearson, Sarah, Hrebien, Heidrun, Gevensleben, Paula Z, Proszek, Michael, Hubank, Margaret, Hills, Jenny, King, Mona, Parmar, Toby, Prout, Laura, Finneran, Jason, Malia, Karen E, Swales, Ruth, Ruddle, Florence I, Raynaud, Alison, Turner, Emma, Hall, Timothy A, Yap, Juanita S, Lopez, and Nicholas C, Turner
- Subjects
Oxazepines ,Phosphatidylinositol 3-Kinases ,Class I Phosphatidylinositol 3-Kinases ,Pyridines ,Receptor, ErbB-2 ,Antineoplastic Combined Chemotherapy Protocols ,Imidazoles ,Humans ,Breast Neoplasms ,Fulvestrant ,Piperazines - Abstract
Cyclin-dependent kinase 4/6 (CDK4/6) and PI3K inhibitors synergize in
- Published
- 2020
16. Preliminary evidence of antitumour activity of Ipatasertib (Ipat) and Atezolizumab (ATZ) in glioblastoma patients (pts) with PTEN loss from the Phase 1 Ice-CAP trial (NCT03673787)
- Author
-
Udai Banerji, Christina Yap, Crescens Diane Tiu, Juanita Lopez, Ruth Riisnaes, Rob Daly, Ben Jenkins, Mateus Crespo, Anna Minchom, J. De-Bono, Suzanne Carreira, Igor Vivanco, Liam Welsh, Alison Turner, Anna Zachariou, Toby Prout, Timothy L. Jones, Bora Gurel, and Nina Tunariu
- Subjects
Cancer Research ,biology ,business.industry ,Immune checkpoint inhibitors ,BNOS 2021 Abstracts ,Disease progression ,medicine.disease ,Ipatasertib ,Tumor excision ,Oncology ,Atezolizumab ,medicine ,Cancer research ,biology.protein ,PTEN ,Neurology (clinical) ,Ice caps ,business ,Glioblastoma - Abstract
Aims Despite improved understanding of effector T-cell trafficking into the central nervous system, initial trials with anti-PD1/PD-L1 immune checkpoint inhibitors (ICIs) have failed to meet their primary endpoints. PTEN loss of function is frequent in GBM and has been correlated with not only poor overall prognosis, but also impaired antitumour responses, including reduced T cell infiltration into tumour and reduced efficacy of ICIs. Ipatasertib is a novel, potent, selective, small-molecule inhibitor of Akt. We have shown that Ipatasertib efficiently depletes FOXP3+ regulatory T cells from the tumour microenvironment (TME) resulting in increased infiltration of effector T cells in solid tumours (Lopez 2020, AACR). We hypothesize that the use of AKT inhibition in PTEN glioblastomas may deplete the TME of suppressive immune cells, and render malignant brain tumours more responsive to ICIs. We present updated data for the combination of Ipat+ATZ in patients with glioblastoma. Method Patients with relapsed WHO grade IV GBM with stable neurological symptoms ≥5 days prior to enrolment, requiring The Ice-CAP A2 cohort assessed safety, pharmacodynamic, and preliminary clinical activity of Ipat (200mg or 400mg OD) + ATZ (1200mg Q3W) in pts with potentially resectable relapsed WHO Grade IV GBM. Pts had a 14-21-day run-in phase of Ipat then surgical tumour resection. Combination Ipat+ATZ commenced post surgery. Patients who declined surgery or who were deemed high risk for surgery proceeded directly to combination. Patients in the expansion cohort B3 commenced directly on Ipat+ATZ at the RP2D of 400mg Ipat with ATZ. Results 16 evaluable recurrent GBM pts were enrolled across two cohorts. Median age 56 yrs (25-71 yrs). Median ECOG PS 1. Median lines of prior therapy 1 (range 1-4). 10 pts had PTEN loss by IHC (H No DLTs, treatment-related (TR) serious adverse events (AEs), or immune-related AEs were observed. Most common TR AEs were G1 diarrhoea (44%), mucositis (17%), rash (28%). Clinical benefit rate (CR, PR and SD> 6 cycles) at clinical cutoff date (23/02/21) in patients with PTEN aberration was 30% (3/10). A 58-year-old man with PTEN loss had MRI at Cycle 5 showing worsening enhancement suggestive of disease progression. Resection of the lesion showed intense lymphocyte infiltration and pathological CR. He is currently on Cycle 22 with no evidence of disease. Two other patients with PTEN loss with radiological stable disease per RANO criteria remain well on study for >6 cycles. Conclusion Combination Ipat+ATZ appears safe and tolerable in GBM pts, with 400mg Ipatasertib OD + 1200mg ATZ Q3W declared as RP2D. Early efficacy signals were detected with PTEN loss being a promising predictive biomarker for response to combination. An expansion cohort enriched with pts with PTEN loss is ongoing.
- Published
- 2021
17. Abstract CT120: Results of the glioblastoma multiforme (GBM) cohort of phase 1 trial Ice-CAP (NCT03673787): Preliminary evidence of antitumour activity of Ipatasertib (Ipa) and Atezolizumab (A) in patients (pts) with PTEN loss
- Author
-
Anna Minchom, Mateus Crespo, Johann S. de Bono, Toby Prout, Igor Vivanco, Crescens Tiu, Suzanne Carreira, Christina Yap, Liam Welsh, Ruth Riisnaes, Ben Jenkins, Alison Turner, Robert Daly, Juanita Lopez, Timothy L. Jones, Udai Banerji, Bora Gurel, Anna Zachariou, Andrea Biondo, and Nina Tunariu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,medicine.disease ,Ipatasertib ,Atezolizumab ,Internal medicine ,Phase (matter) ,Cohort ,medicine ,biology.protein ,PTEN ,In patient ,Ice caps ,business ,Glioblastoma - Abstract
Background: Hyperactivation of the PI3K/AKT pathway correlates with impaired antitumour response, including reduced T cell infiltration into tumour and reduced efficacy of immune checkpoint inhibitors (ICIs). PTEN loss of function, often observed in GBM, may contribute to refractoriness of ICIs in this disease. Methods: The Ice-CAP A2 cohort assessed safety, pharmacodynamic, and preliminary clinical activity of Ipa (200mg or 400mg OD) + A (1200mg Q3W) in pts with potentially resectable relapsed WHO Grade IV GBM. Key inclusion criteria were stable neurological symptoms ≥5 days prior to enrolment, steroid requirement 12wks, both on 400mg Ipa. A 58-year-old man with PTEN loss had MRI at Cycle 5 showing worsening enhancement suggestive of disease progression. Resection of the lesion showed intense lymphocyte infiltration and pathological CR. He is currently on Cycle 18 with no evidence of disease. Conclusion: Combination Ipa+A appears safe and tolerable in GBM pts, with 400mg Ipa OD + 1200mg A Q3W declared as RP2D. PTEN loss may be a promising predictive biomarker for response to combination. An expansion cohort enriched with pts with PTEN loss is ongoing. Cytokine and FACS data will be presented at AACR Table 1.Clinical Benefit Rate of glioblastoma patients stratified according to PTEN aberrationsPTEN statusnBest responseClinical Benefit RatePTEN loss on IHC (H12 wks, ongoingb3 PDPTEN aberration on NGS but PTEN protein expression pending11 PDcPTEN loss of heterozygozity on PCR11 PDWild type PTEN on NGS or IHC (H≥30)33 PDLegend: IHC = immunohistochemistry; NGS = next generation sequencing; PCR = polymerase chain reaction;pCR = pathologic complete response; SD = stable disease; PD = progressive diseaseaExceptional responder with PTEN H=5 on IHC and splice site 75_79+2delGACCTGT on NGSb PTENY68*; c PTENQ298* Citation Format: Crescens Tiu, Andrea Biondo, Liam C. Welsh, Timothy L. Jones, Anna Zachariou, Toby Prout, Alison J. Turner, Robert Daly, Igor Vivanco, Christina Yap, Ben Jenkins, Mateus Crespo, Ruth Riisnaes, Suzanne Carreira, Bora Gurel, Nina Tunariu, Anna Minchom, Udai Banerji, Johann S. de Bono, Juanita S. Lopez. Results of the glioblastoma multiforme (GBM) cohort of phase 1 trial Ice-CAP (NCT03673787): Preliminary evidence of antitumour activity of Ipatasertib (Ipa) and Atezolizumab (A) in patients (pts) with PTEN loss [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT120.
- Published
- 2021
18. Abstract CT129: HyPeR: A phase 1, dose escalation and expansion trial of guadecitabine (SGI-110), a second-generation hypomethylating agent in combination with pembrolizumab (MK3475) in patients with refractory solid tumors
- Author
-
Toby Prout, Ricardo Morilla, Mark Van de Velde, Ruth Riisnaes, Malaka Ameratunga, Johann S. de Bono, Claudia Bertan, Abhijit Pal, Ines Figueiredo, Christina Yap, Mateus Crespo, Melek Akay, Susana Miranda, Nahal Masrour, Robert M Brown, Wei Yuan, Chloe Baker, Ben Jenkins, Sanjena Mithra, Anna Ferreira, Bora Gurel, Juanita Lopez, Anna Zachariou, Sofia Levva, Nina Tunariu, Reece Caldwell, Alison Turner, Udai Banerji, Joo-Ern Ang, Suzanne Carreira, Dionysis Papadatos-Pastos, Anna Minchom, George Seed, Rita Pereira, and Mona Parmar
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Tumor-infiltrating lymphocytes ,Alpha interferon ,Cancer ,Pembrolizumab ,Neutropenia ,medicine.disease ,Gastroenterology ,Oncology ,Hypomethylating agent ,Internal medicine ,Biopsy ,medicine ,business ,Febrile neutropenia - Abstract
Background: Methylation is reported to support cancer immune tolerance. We conducted a phase 1 dose-escalation trial [NCT02998567] of combination guadecitabine (G; DNA hypomethylating agent) and pembrolizumab (P) in patients (pts) with advanced cancers. We hypothesized that G can normalize the expression of epigenetically suppressed immune genes, increase interferon producing tumor-infiltrating lymphocytes (TILs), and enhance the anticancer activity of P. Methods: In dose escalation (Es), pts received G (45 mg/m2 or 30 mg/m2, administered SC on days 1-4) with P (200 mg, administered IV starting from cycle 2 onwards) as outpatient Q3W; in expansion (Ex), the RP2D of G (30 mg/m2) with P (200 mg) Q3W was administered. Pre-treatment and on-treatment tumor biopsies were evaluated for PD-L1 expression, tumor infiltrating lymphocytes, gene expression by RNAseq and methylome studies. Longitudinal analyses of peripheral blood CD3, CD4 and CD8 lymphocytes by flow cytometry were performed. Results: Overall, 34 pts (Es, n = 14; Ex, n = 20) were evaluable for safety. The most common treatment-related adverse events (TRAEs) were neutropenia (n = 21), fatigue (n = 6) and thrombocytopenia (n = 3), diarrhea (n = 2). G3+ TRAEs were neutropenia (n = 14), febrile neutropenia (n = 4), raised ALP (n = 1), raised AST (n=1), colitis (n = 1), diarrhoea (n = 1) and lung infection (n = 1). Two DLTs (neutropenia, febrile neutropenia) were reported at G 45mg/m2 with none reported at G 30mg/m2. There were no treatment-related deaths. In total, 28 pts (Es, n = 12; Ex, n = 16) were evaluable for antitumor activity studies (≥2 scans); ORR (CR+PR) and DCR (CR+PR+SD) were 3% and 57%; 10/15 pts with non-small cell lung cancer (13 pts resistant/refractory to PD-1/PD-L1 targeting agents) were evaluable, with a DCR of 80% and 5 pts having DCR > 6 months with 8 pts remaining on study treatment. Overall, 25 paired biopsies were obtained. Using LINE1 sequences to study global methylation, both tumor biopsies and peripheral blood showed reduced methylation post-G treatment. Preliminary data on tumor-infiltrating lymphocytes assessed by multicolor immunofluorescence in 9 paired biopsies showed a numerical increase in median values of T-helper (CD4+FOXP3-) (10.20 to 19.70, p = 0.5469), T-regulatory (CD4+FOXP3+) (5.1 to 6.7, p=0.8438), and T-cytotoxic (CD8+) cell densities (2.7 to 7.4, p=0.6523) . Comparing with matched pre-treatment, on treatment tumor had numerical increases in interferon alpha and gamma response pathway activation in serial biopsy RNAseq analyses but did not reach significance. Conclusions: G plus P resulted in no unexpected toxicities with evidence suggestive of biological and anti-cancer activity. Citation Format: Dionysis Papadatos-Pastos, Abhijit Pal, Melek Akay, Malaka Ameratunga, Sanjena Mithra, Joo-Ern Ang, Sofia Levva, Reece Caldwell, Ruth Riisnaes, Mateus Crespo, Wei Yuan, George Seed, Bora Gurel, Ines Figueiredo, Rita Pereira, Susana Miranda, Anna Ferreira, Suzanne Carreira, Claudia Bertan, Chloe Baker, Ricardo Morilla, Robert Brown, Nahal Masrour, Toby Prout, Anna Zachariou, Alison Turner, Mona Parmar, Mark Van de Velde, Ben Jenkins, Christina Yap, Nina Tunariu, Udai Banerji, Juanita Lopez, Anna Minchom, Johann De Bono. HyPeR: A phase 1, dose escalation and expansion trial of guadecitabine (SGI-110), a second-generation hypomethylating agent in combination with pembrolizumab (MK3475) in patients with refractory solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT129.
- Published
- 2020
19. Abstract CT140: Proof-of-concept evidence of immune modulation by blockade of the phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway in the phase I dose escalation study of Ipatasertib (Ipa) in combination with atezolizumab (A) in patients (pts) with advanced solid tumors (Ice-CAP)
- Author
-
Nina Tunariu, Anna Zachariou, Johann S. de Bono, Toby Prout, Crescens Diane Tiu, Ben Jenkins, Hannah Badham, Ricardo Morilla, Anna Minchom, Juanita Lopez, Ruth Riisnaes, Ines Figueiredo, Karen E Swales, Mateus Crespo, Bora Gurel, Christina Yap, Rob Daly, Mariana Scaranti, Alison Turner, Igor Vivanco, Udai Banerji, Mona Parmar, Malaka Ameratunga, Rita Pereira, Andrea Biondo, and Wei Yuan
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,FOXP3 ,Phases of clinical research ,Rash ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Cytokine ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,medicine.symptom ,business ,PI3K/AKT/mTOR pathway - Abstract
Background: Hyperactivation of the PI3K/AKT pathway correlates with impaired anti-tumor responses, including reduced T cell infiltration into tumor, and reduced efficacy of immune checkpoint inhibitors. Blockade of this pathway synergizes with PD-L1/PD-1 axis blockade preclinically. Methods: This Phase I clinical trial (NCT03673787) assessed the safety, pharmacodynamic, and preliminary clinical activity of Ipa (200mg or 400mg OD) given in combination with A 1200mg q3 wk in refractory pts. Serial paired blood and tumor samples were analysed to interrogate the effect of Ipa on the tumor micro-environment and host immune system prior to the addition of the immune check point inhibitor, A. Results: 18 adult pts were treated in dose escalation. Median age 49 yrs. All pts had ECOG PS 0-1 and median 7 prior therapies. Most common TRAEs (>15%) were mild Gr1-2 diarrhea (56%), rash (50%), fatigue (33%), nausea (33%), raised ALT/AST (33%), headache (28%) and arthralgia (22%). 1 pt had G2 systemic immune activation; 2 pts had G3 rash, both rapidly reversible. 1 DLT of G3 raised ALT seen at 200mg (1 DLT/9 evaluable pts) but none at 400mg (0 DLT/6). Of 14 RECIST evaluable patients, there were 2 confirmed PRs, and 5 SD (clinical benefit rate 50%). Reductions of CD4+FOXP3+ Tregs in tumor microenvironment were seen after 2wks of single agent Ipa, regardless of PIK3/AKT somatic mutation status (Table 1). Responding pts had a >400% median increase in intra-tumoral CD8+ Teff cell infiltration, effectively switching from a desert phenotype to an inflamed phenotype. Paired changes in FACS, transcriptome and cytokine will also be presented.Conclusions: The RP2D of Ipa 400mg OD combination with A was well tolerated with early efficacy signals. Further biomarker work is ongoing and will be evaluated in expansion cohorts. Table 1:Changes in immune cell populations as assessed by multicolour Immunofluorescence in paired biopsies of breast/gynae patients, % change in cell number/mm2 from baseline (median [min,max$])&Post 2 weeks single agent Ipatasertib(n=9)Post 1 cycle of combination Ipatasertib and Atezolizumab(n=7)CD4+FOXP3+Tregs cellsCD 8+ Teff cellsCD4+FOXP3+Tregs cellsCD 8+ Teff cellsIntra-tumourstromaIntra-tumourstromaIntra-tumourstromaIntra-tumourstromaAll patients-23.9*[-89.7, BL0]-30.0*[-91.6, BL0]-37.7*[-84.4, -24.5]-28.4[-92.4, 259.8]335.9[-44.0,BL0]45.4[-51.0, BL0]59.6[-60.6,493.3]64.7[-51.7,293.3]Stratified by somatic PI3K/AKT/PTEN mutational statusPathogenic mutations (mt)11.1[-82.2, BL0]#-10.7[-91.6, BL0]Φnsnsnsns-30.5[-60.6,-0.5]11.3[-51.7,50.0]Wildtype (wt)-63.1[-89.7,19.0]#-47.5[-77.0,11.1]Φnsnsnsns426.5[59.6,493.3]126.7[79.4,293.3]Stratified by responseResponders (PR + SD>4 cycles). 1 ER+ HER2+ breast cancer (wt), 1 ER+ HER2- breast cancer (wt)459.9[426.5,493.3]@103.1[79.4,126.7]Non-responders (PD at 4 cycles) 1 cervical cancer, 4 ER+ breast cancer-0.5[-60.6, 59.6]@30.6[-51.7,293.3]*significant change (p≤0.05; Wilcoxon sign-rank test) from baseline, $maximum values denoted by BL0indicate that the baseline value was zero, and so percentage change from baseline is not defined. For the analysis, the baseline value has been replaced by a nominal value of 0.1 so that a large percentage increase is associated with these cases. Note that these large percentage increases do not affect the non-parametric statistical tests used.#no significant difference in distribution of reduction in intra-tumoural CD4+ FOXP3+Tregsbetween pts with pathogenic mutations in PI3K/AKT and those without (p=0.30; Wilcoxon rank-sum test)Φno significant difference in distribution of reduction in stromal CD4+FOXP3+Tregsbetween pts with pathogenic mutations in PI3K/AKT and those without (p=0.44; Wilcoxon rank-sum test) @ difference between responders and non-responders p=0.083; Wilcoxon rank-sum test)mt pathogenic mutations in PI3K/AKT and PTEN as per COSMIC database present in tumour or PTEN loss by IHC. wt no pathogenic mutations in PI3K/AKT and PTEN as per COSMIC database detected in tumour and intact PTEN expression by IHC. &exploratory analyses with no adjustment for multiple testing Citation Format: Juanita S. Lopez, Andrea Biondo, Crescens Tiu, Mariana Scaranti, Malaka Ameratunga, Anna Zachariou, Alison Turner, Nina Tunariu, Toby Prout, Mona Parmar, Hannah Badham, Karen Swales, Wei Yuan, Ricardo Morilla, Mateus Crespo, Rob Daly, Ines Figueiredo, Bora Gurel, Rita Pereira, Ruth Riisnaes, Igor Vivanco, Anna Minchom, Ben Jenkins, Christina Yap, Udai Banerji, Johann De Bono. Proof-of-concept evidence of immune modulation by blockade of the phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway in the phase I dose escalation study of Ipatasertib (Ipa) in combination with atezolizumab (A) in patients (pts) with advanced solid tumors (Ice-CAP) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT140.
- Published
- 2020
20. A phase I trial a FR alpha targeted thymidylate synthase inhibitor CT900 exploring four schedules of treatment in expansion cohorts of patients with high-grade serous ovarian cancer
- Author
-
A. Biondo, Ed Ainscow, Bora Gurel, Ben Jenkins, Udai Banerji, Natalie Cook, Alvaro Ingles Russo, Anna Minchom, Toby Prout, Ruth Riisnaes, Stuart McIntosh, Bristi Basu, Martin Little, Emma Hall, Juanita Lopez, Susana Banerjee, Ionut-Gabriel Funingana, Mona Parmar, Johann S. de Bono, and Nina Tunariu
- Subjects
Folate Receptor Alpha ,Cancer Research ,biology ,business.industry ,Alpha (ethology) ,Small molecule ,Thymidylate synthase ,Oncology ,Thymidylate synthase inhibitor ,Serous ovarian cancer ,Cancer research ,biology.protein ,Medicine ,Cytotoxicity ,business - Abstract
6043 Background: CT900 (BTG945/ONX-0801) is a novel small molecule that binds to folate receptor alpha (FRα), is internalized and causes cytotoxicity by thymidylate synthase inhibition. Methods: The aims of the expansion cohorts were to determine toxicity, response rates and correlation of the response to FRα expression in patients with HGSOC (NCT02360345). Four expansion cohorts were studied which included: schedule A (6 mg/m2/q every 2 weeks), schedule B (12 mg/m2/q every 2 weeks), schedule C (12 mg/m2/q every 2 weeks with 12 mg dexamethasone IV and 8 mg of dexamethasone for 2 days) and schedule D (12 mg/m2/q every 3 weeks). Response rates were assessed by RECIST V1.1 and GCIG CA125 response criteria. Patients who were withdrawn for reasons other than toxicity within 8 weeks (cohorts A, B, C) and 12 weeks (cohort D) were not assessable for efficacy. FRα expression was quantified using immunohistochemistry. Results: A total of 67 patients were treated in the 4 cohorts (14, 25, 15 and 13 for cohorts A, B, C and D). The median age was 62 (IQR 57 - 68) and the median lines of previous treatment was 5 (range 1 to 13). A majority of patients were platinum resistant. The most common toxicities across all expansion cohorts were: fatigue (51%), nausea (36%), anemia (27%), fever (25%), AST elevation (21%), most of which were grade 1 - 2. Toxicity of special interest included radiological changes of pneumonitis and was 15% in all cohorts (7%, 16%, 27% and 8% in cohorts A, B, C and D, respectively). These changes were grade 1 - 2 in all but one case. RECIST response rates in evaluable patients across the different cohorts were: A 1/8 (13%), B 6/21 (29%), C 5/12 (42%) and D 2/12 (17%). FRα expression in archival tumor tissue was measured in 59/67 patients. Expression was found to be high/medium in 43/59 (73%), low in 7/59 (12%) and negative/very low in 9/59 (15%). In patients with high/medium FRα expression, the RECIST response rates in different cohorts were: A 0/9 (0%), B 6/16 (38%), C 4/12 (33%) and D 1/6 (17%). The CA125 response rate in all patients within cohort B was 13/25 (52%) and 10/16 (63%) in patients with high/medium FRα expression. Conclusions: CT900 has shown clinical activity in patients with heavily pre-treated platinum-resistant, high/medium FRα expressing HGSOC. Based on toxicity and efficacy, the schedule of 12 mg/m2/q2 weekly (schedule B) is the recommended phase II dose for further evaluation in patients with relapsed high/medium FRα expressing HGSOC. Clinical trial information: NCT02360345.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.