137 results on '"Jean-François Baurain"'
Search Results
2. Hysteroscopic management of molar pregnancy: A series of 36 cases
- Author
-
Matthieu de Codt, Pascale Jadoul, Mathieu Luyckx, Jean-Luc Squifflet, Marie-Madeleine Dolmans, Charlotte Maillard, Jean-François Baurain, Etienne Marbaix, and Amandine Gerday
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Hydatidiform Mole (HM) is the most common form of gestational trophoblastic disease. Dilatation and curettage is the classical treatment of this affection. Hysteroscopic resection (HsR) is an alternative for the treatment of intra-uterine pathology. Objective: To describe the feasibility of HsR for the management of HM. Result: Case series of patients who had a complete or partial HM confirmed by histological examination of the trophoblastic tissue resected by operative hysteroscopy between 2007 and 2019. After approval of our ethics committee, we evaluated 36 patients who underwent hysteroscopic resection for molar pregnancy. Histological analysis showed partial HM in 28 patients (77.8%) and complete HM in 8 (22.2%). Main surgical complications were uterine perforation in one patient and glycine resorption in 10 patients with two cases of hyponatremia corrected by standard treatment. We performed an ultrasound control 1 month after the intervention in 19 patients (52.8%) as they had slow decrease of HCG or bleeding complaints and found retained product of conception (RPOC) in six patients (16.7%). Conclusion: This first report on a small number of patients demonstrate that hysteroscopic resection is a feasible procedure for the management of molar pregnancy. Direct visualization of the procedure helps the surgeon to control the resection. Further studies are mandatory to compare this technique with D&C in term of RPOC and fertility outcomes as it remains the standard treatment.
- Published
- 2023
- Full Text
- View/download PDF
3. Hyperpolarized 13C-Pyruvate to Assess Response to Anti-PD1 Immune Checkpoint Inhibition in YUMMER 1.7 Melanoma Xenografts
- Author
-
Chantale Farah, Marie-Aline Neveu, Caroline Bouzin, Zorica Knezevic, Bernard Gallez, Eleonora Leucci, Jean-François Baurain, Lionel Mignion, and Bénédicte F. Jordan
- Subjects
melanoma ,tumor metabolism ,immunotherapy ,response biomarkers ,13C-MRS ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
There is currently no consensus to determine which advanced melanoma patients will benefit from immunotherapy, highlighting the critical need to identify early-response biomarkers to immune checkpoint inhibitors. The aim of this work was to evaluate in vivo metabolic spectroscopy using hyperpolarized (HP) 13C-pyruvate and 13C-glucose to assess early response to anti-PD1 therapy in the YUMMER1.7 syngeneic melanoma model. The xenografts showed a significant tumor growth delay when treated with two cycles of an anti-PD1 antibody compared to an isotype control antibody. 13C-MRS was performed in vivo after the injection of hyperpolarized 13C-pyruvate, at baseline and after one cycle of immunotherapy, to evaluate early dynamic changes in 13C-pyruvate–13C-lactate exchange. Furthermore, ex vivo 13C-MRS metabolic tracing experiments were performed after U-13C-glucose injection following one cycle of immunotherapy. A significant decrease in the ratio of HP 13C-lactate to 13C-pyruvate was observed in vivo in comparison with the isotype control group, while there was a lack of change in the levels of 13C lactate and 13C alanine issued from 13C glucose infusion, following ex vivo assessment on resected tumors. Thus, these results suggest that hyperpolarized 13C-pyruvate could be used to assess early response to immune checkpoint inhibitors in melanoma patients.
- Published
- 2023
- Full Text
- View/download PDF
4. HOXA1 Is an Antagonist of ERα in Breast Cancer
- Author
-
Magali Belpaire, Bruno Ewbank, Arnaud Taminiau, Laure Bridoux, Noémie Deneyer, Damien Marchese, Gipsi Lima-Mendez, Jean-François Baurain, Dirk Geerts, and René Rezsohazy
- Subjects
HOX proteins ,estrogen receptor ,NF-κB ,endocrine therapy resistance ,PBX ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Breast cancer is a heterogeneous disease and the leading cause of female cancer mortality worldwide. About 70% of breast cancers express ERα. HOX proteins are master regulators of embryo development which have emerged as being important players in oncogenesis. HOXA1 is one of them. Here, we present bioinformatic analyses of genome-wide mRNA expression profiles available in large public datasets of human breast cancer samples. We reveal an extremely strong opposite correlation between HOXA1 versus ER expression and that of 2,486 genes, thereby supporting a functional antagonism between HOXA1 and ERα. We also demonstrate in vitro that HOXA1 can inhibit ERα activity. This inhibition is at least bimodal, requiring an intact HOXA1 DNA-binding homeodomain and involving the DNA-binding independent capacity of HOXA1 to activate NF-κB. We provide evidence that the HOXA1-PBX interaction known to be critical for the transcriptional activity of HOXA1 is not involved in the ERα inhibition. Finally, we reveal that HOXA1 and ERα can physically interact but that this interaction is not essential for the HOXA1-mediated inhibition of ERα. Like other HOX oncoproteins interacting with ERα, HOXA1 could be involved in endocrine therapy resistance.
- Published
- 2021
- Full Text
- View/download PDF
5. Combined HP 13C Pyruvate and 13C-Glucose Fluxomic as a Potential Marker of Response to Targeted Therapies in YUMM1.7 Melanoma Xenografts
- Author
-
Chantale Farah, Marie-Aline Neveu, Caner Yelek, Caroline Bouzin, Bernard Gallez, Jean-François Baurain, Lionel Mignion, and Bénédicte F. Jordan
- Subjects
melanoma ,tumor metabolism ,targeted therapy ,BRAF and MEK inhibition ,13C-MRS ,markers of response ,Biology (General) ,QH301-705.5 - Abstract
A vast majority of BRAF V600E mutated melanoma patients will develop resistance to combined BRAF/MEK inhibition after initial clinical response. Resistance to targeted therapy is described to be accompanied by specific metabolic changes in melanoma. The aim of this work was to evaluate metabolic imaging using 13C-MRS (Magnetic Resonance Spectroscopy) as a marker of response to BRAF/MEK inhibition in a syngeneic melanoma model. Tumor growth was significantly delayed in mice bearing YUMM1.7 melanoma xenografts treated with the BRAF inhibitor vemurafenib, and/or with the MEK inhibitor trametinib, in comparison with the control group. 13C-MRS was performed in vivo after injection of hyperpolarized (HP) 13C-pyruvate, at baseline and 24 h after treatment, to evaluate dynamic changes in pyruvate-lactate exchange. Furthermore, ex vivo 13C-MRS steady state metabolic tracing experiments were performed after U-13C-glucose or 5-13C-glutamine injection, 24 h after treatment. The HP 13C-lactate-to-pyruvate ratio was not modified in response to BRAF/MEK inhibition, whereas the production of 13C-lactate from 13C-glucose was significantly reduced 24 h after treatment with vemurafenib, trametinib, or with the combined inhibitors. Conversely, 13C-glutamine metabolism was not modified in response to BRAF/MEK inhibition. In conclusion, we identified 13C-glucose fluxomic as a potential marker of response to BRAF/MEK inhibition in YUMM1.7 melanoma xenografts.
- Published
- 2022
- Full Text
- View/download PDF
6. Highly Aggressive Metastatic Melanoma Cells Unable to Maintain Telomere Length
- Author
-
Nikenza Viceconte, Marie-Sophie Dheur, Eva Majerova, Christophe E. Pierreux, Jean-François Baurain, Nicolas van Baren, and Anabelle Decottignies
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Unlimited replicative potential is one of the hallmarks of cancer cells. In melanoma, hTERT (telomerase reverse transcriptase) is frequently overexpressed because of activating mutations in its promoter, suggesting that telomerase is necessary for melanoma development. We observed, however, that a subset of melanoma metastases and derived cell lines had no telomere maintenance mechanism. Early passages of the latter displayed long telomeres that progressively shortened and fused before cell death. We propose that, during melanoma formation, oncogenic mutations occur in precursor melanocytes with long telomeres, providing cells with sufficient replicative potential, thereby bypassing the need to re-activate telomerase. Our data further support the emerging idea that long telomeres promote melanoma formation. These observations are important when considering anticancer therapies targeting telomerase. : Viceconte et al. report that metastatic cutaneous melanoma cells display long telomeres and do not always have an active telomere maintenance mechanism, showing that the ability to maintain telomeres is not an absolute requirement for metastasis formation. These observations are important in the context of anti-cancer therapies targeting telomerase. Keywords: telomeres, telomerase, melanoma, ever-shorter telomeres, immortalization
- Published
- 2017
- Full Text
- View/download PDF
7. Fertility Sparing Treatment in Patients With Early Stage Endometrial Cancer, Using a Combination of Surgery and GnRH Agonist: A Monocentric Retrospective Study and Review of the Literature
- Author
-
Stéphanie Tock, Pascale Jadoul, Jean-Luc Squifflet, Etienne Marbaix, Jean-François Baurain, and Mathieu Luyckx
- Subjects
endometrial adenocarcinoma ,conservative treatment ,atypical endometrial hyperplasia ,GnRH agonist ,fertility-sparing ,Medicine (General) ,R5-920 - Abstract
Objectives: To evaluate the efficacy and safety of gonadotropin-releasing hormone (GnRH) agonist after endometrial resection in women suffering early stage endometrial carcinoma (EC) and/or endometrial intra-epithelial neoplasia (EIN).Design: A retrospective review of clinical files between January 1999 and December 2016.Setting: University hospital.Patients: Eighteen women younger than 41 years with grade 1 endometrial carcinoma (G1EC) and/or Endometrial intra-epithelial neoplasia (EIN). Interventions: All patients received GnRH agonist for 3 months after an endometrial resection combined with a laparoscopy to exclude concomitant ovarian tumor and/or other extra-uterine disease. The patient underwent a follow-up of 3 months interval with endometrial sampling by hysteroscopy.Main Outcome Measure(s): The recurrence rate and the pregnancy rate after fertility sparing treatment.Results: We identified 9 patients with EIN (50%), 7 patients with G1EC (38.9%), 1 with combined histology (5.5%), and 1 with G2EC (5.5%). After a median follow-up of 40.7 months, 12 patients conserved their uterus (66.7%), and 8 (53.3%) patients were pregnant with a total of 14 pregnancies among those who tried to become pregnant. We observed a complete response rate in 12 patients (66.7%) but 3 of these patients relapsed (25%). We also found a stable disease in 6 patients (33.3%).Conclusions: Compared with other fertility sparing treatments, GnRH agonist after surgery is an effective fertility-sparing strategy for women with EIN and/or G1EC. We recommend hysterectomy once a family has been completed even if the literature does not clearly lead to radical surgery.
- Published
- 2018
- Full Text
- View/download PDF
8. Selective use of sequential digital dermoscopy imaging allows a cost reduction in the melanoma detection process: a belgian study of patients with a single or a small number of atypical nevi.
- Author
-
Isabelle Tromme, Brecht Devleesschauwer, Philippe Beutels, Pauline Richez, Nicolas Praet, Laurine Sacré, Liliane Marot, Pascal Van Eeckhout, Ivan Theate, Jean-François Baurain, Julien Lambert, Catherine Legrand, Luc Thomas, and Niko Speybroeck
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Dermoscopy is a technique which improves melanoma detection. Optical dermoscopy uses a handheld optical device to observe the skin lesions without recording the images. Sequential digital dermoscopy imaging (SDDI) allows storage of the pictures and their comparison over time. Few studies have compared optical dermoscopy and SDDI from an economic perspective. OBJECTIVE: The present observational study focused on patients with one-to-three atypical melanocytic lesions, i.e. lesions considered as suspicious by optical dermoscopy. It aimed to calculate the "extra-costs" related to the process of melanoma detection. These extra-costs were defined as the costs of excision and pathology of benign lesions and/or the costs of follow-up by SDDI. The objective was to compare these extra-costs when using optical dermoscopy exclusively versus optical dermoscopy with selective use of SDDI. METHODS: In a first group of patients, dermatologists were adequately trained in optical dermoscopy but worked without access to SDDI. They excised all suspicious lesions to rule out melanoma. In a second group, the dermatologists were trained in optical and digital dermoscopy. They had the opportunity of choosing between immediate excision or follow-up by SDDI (with delayed excision if significant change was observed). The comparison of extra-costs in both groups was made possible by a decision tree model and by the division of the extra-costs by the number of melanomas diagnosed in each group. Belgian official tariffs and charges were used. RESULTS: The extra-costs in the first and in the second group were respectively €1,613 and €1,052 per melanoma excised. The difference was statistically significant. CONCLUSIONS: Using the Belgian official tariffs and charges, we demonstrated that the selective use of SDDI for patients with one-to-three atypical melanocytic lesions resulted in a significant cost reduction.
- Published
- 2014
- Full Text
- View/download PDF
9. A Synopsis of Serum Biomarkers in Cutaneous Melanoma Patients
- Author
-
Pierre Vereecken, Frank Cornelis, Nicolas Van Baren, Valérie Vandersleyen, and Jean-François Baurain
- Subjects
Dermatology ,RL1-803 - Abstract
Many serum biomarkers have been evaluated in melanoma but their clinical significance remains a matter of debate. In this paper, a review of the serum biomarkers for melanoma will be detailed and will be discussed from the point of view of their practical usefulness. The expression of biomarkers can be detected intracellularly or on the cell membrane of melanoma cells or noncancer cells in association with the melanoma. Some of these molecules can then be released extracellularly and be found in body fluids such as the serum. Actually, with the emergence of new targeted therapies for cancer and the increasing range of therapeutic options, the challenge for the clinician is to assess the unique risk/response ratio and the prognosis for each patient. New serum biomarkers of melanoma progression and metastatic disease are still awaited in order to provide efficient rationale for followup and treatment choices. LDH as well as S100B levels have been correlated with poor prognosis in AJCC stage III/IV melanoma patients. However, the poor sensitivity and specificity of those markers and many other molecules are serious limitations for their routine use in both early (AJCC stage I and II) and advanced stages of melanoma (AJCC stage III and IV). Microarray technology and proteomic research will surely provide new candidates in the near future allowing more accurate definition of the individual prognosis and prediction of the therapeutic outcome and select patients for early adjuvant strategies.
- Published
- 2012
- Full Text
- View/download PDF
10. Prospective evaluation of the tolerability and efficacy of niraparib dosing based on baseline body weight and platelet count: Results from the PRIMA/ENGOT‐OV26/GOG‐3012 trial
- Author
-
Mansoor R. Mirza, Antonio González‐Martín, Whitney S. Graybill, David M. O’Malley, Lydia Gaba, Oi Wah Stephanie Yap, Eva M. Guerra, Peter G. Rose, Jean‐François Baurain, Sharad A. Ghamande, Hannelore Denys, Emily Prendergast, Carmela Pisano, Philippe Follana, Klaus Baumann, Paula M. Calvert, Jacob Korach, Yong Li, Izabela A. Malinowska, Divya Gupta, and Bradley J. Monk
- Subjects
poly(ADP-ribose) polymerase inhibitors/adverse effects ,Cancer Research ,ovarian cancer ,Oncology ,Medicine and Health Sciences ,individualized starting dose ,niraparib ,poly(ADP-ribose) polymerase inhibitors/therapeutic use - Abstract
Background: The PRIMA/ENGOT-OV26/GOG-3012 (NCT02655016) trial was amended to prospectively evaluate the safety and efficacy of an individualized starting dose (ISD) regimen of niraparib for first-line maintenance therapy in patients with newly diagnosed advanced ovarian cancer. Methods: In the phase 3 PRIMA trial, patients with newly diagnosed advanced ovarian cancer with a complete/partial response to first-line platinum-based chemotherapy (N = 733) were initially treated with a fixed starting dose (FSD) regimen of 300 mg every day. Subsequently, the protocol was amended so newly enrolled patients received an ISD: 200 mg every day in patients with baseline body weight < 77 kg or baseline platelet count < 150,000/mu L, and 300 mg every day in all other patients. Efficacy and safety outcomes were assessed by starting dose. Results: Overall, 475 (64.8%) patients were assigned to an FSD (niraparib, n = 317; placebo, n = 158) and 258 (35.2%) were assigned to an ISD (niraparib, n = 170; placebo, n = 88). Efficacy in patients who received FSD or ISD was similar for the overall (FSD hazard ratio [HR], 0.59 [95% CI, 0.46-0.76] vs. ISD HR, 0.69 [95% CI, 0.48-0.98]) and the homologous recombination-deficient (FSD HR, 0.44 [95% CI, 0.30-0.64] vs. ISD HR, 0.39 [95% CI, 0.22-0.72]) populations. In patients with low body weight/platelet count, rates of grades =3 and 4 hematologic treatment-emergent adverse events, dose interruptions, and dose reductions were lower for those who received ISD than for those who received FSD. Conclusions: In PRIMA, similar dose intensity, similar efficacy, and improved safety were observed with the ISD compared with the FSD regimen.
- Published
- 2023
11. Supplementary Fig. S1 from A Short Treatment with Galactomannan GM-CT-01 Corrects the Functions of Freshly Isolated Human Tumor–Infiltrating Lymphocytes
- Author
-
Pierre van der Bruggen, Pierre J. Courtoy, Patrick Van Der Smissen, Jean-François Baurain, Kris Thielemans, Javier Carrasco, Jean-Luc Squifflet, Vincent Stroobant, Grégoire Wieërs, René Bigirimana, and Nathalie Demotte
- Abstract
PDF file 98K, Influence of GM-CT-01 on T cell and melanoma cell growth
- Published
- 2023
12. Supplementary Table S1 from A Short Treatment with Galactomannan GM-CT-01 Corrects the Functions of Freshly Isolated Human Tumor–Infiltrating Lymphocytes
- Author
-
Pierre van der Bruggen, Pierre J. Courtoy, Patrick Van Der Smissen, Jean-François Baurain, Kris Thielemans, Javier Carrasco, Jean-Luc Squifflet, Vincent Stroobant, Grégoire Wieërs, René Bigirimana, and Nathalie Demotte
- Abstract
PDF file 105K, GM-CT-01 boosts secretion of various cytokines by TIL
- Published
- 2023
13. Data from A Short Treatment with Galactomannan GM-CT-01 Corrects the Functions of Freshly Isolated Human Tumor–Infiltrating Lymphocytes
- Author
-
Pierre van der Bruggen, Pierre J. Courtoy, Patrick Van Der Smissen, Jean-François Baurain, Kris Thielemans, Javier Carrasco, Jean-Luc Squifflet, Vincent Stroobant, Grégoire Wieërs, René Bigirimana, and Nathalie Demotte
- Abstract
Purpose: Several galectins are released by tumor cells and macrophages and accumulate in the tumor microenvironment. Galectin-1 and -3 were found to bind to glycosylated receptors at the surface of tumor-infiltrating lymphocytes (TIL), forming glycoprotein–galectin lattices that could reduce the motility and therefore the functionality of surface molecules. In contrast to blood T cells, human TIL show defective IFN-γ secretion upon ex vivo stimulation. We have previously shown that extracellular galectin-3 participates in the impairment of TIL functions. Indeed, disruption of glycoprotein–galectin-3 lattices using anti-galectin-3 antibodies, or N-acetyllactosamine as a competing sugar, boosted cytokine secretion by TIL. Here we have tested a clinical grade galectin antagonist: GM-CT-01, a galactomannan obtained from guar gum reported to be safe in more than 50 patients with cancer.Experimental Design: TIL were isolated from human tumor ascites, treated for 2 to 20 hours with galectin antagonists and tested for function.Results: We found that GM-CT-01 boosts cytotoxicity of CD8+ TIL and their IFN-γ secretion in a dose-dependent manner. Treating TIL obtained from patients with various cancers, during a few hours, resulted in an increased IFN-γ secretion in up to 80% of the samples.Conclusions: These observations pave the way for investigating the potential benefit of this galectin antagonist in patients with cancer, alone or combined with cancer vaccination, in order to correct in vivo impaired functions of TIL. Clin Cancer Res; 20(7); 1823–33. ©2014 AACR.
- Published
- 2023
14. Supplementary Figure 3 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 131K, Procedure for the preparation of immunoglobulin VDJC1 libraries
- Published
- 2023
15. Supplementary Table 2 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 37K, Main characteristics of patients and tumor samples
- Published
- 2023
16. Supplementary Figure 1 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 418K, Immunohistological detection of the indicated antigens in adjacent cryosections of cutaneous melanoma metastasis T9
- Published
- 2023
17. Supplementary Figure 4 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 4.3MB, Immunohistological detection of the indicated antigens in adjacent cryosections of primary melanoma T33
- Published
- 2023
18. Supplementary Table 1 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 49K, List and principal characteristics of antibodies and oligonucleotide primers used in the study
- Published
- 2023
19. Supplementary Figure 2 from Neogenesis of Lymphoid Structures and Antibody Responses Occur in Human Melanoma Metastases
- Author
-
Nicolas van Baren, Pierre G. Coulie, Thierry Boon, Marguerite Stas, Joost van den Oord, Ivan Théate, Jean-François Baurain, Teofila Seremet, Marjorie Mercier, and Arcadi Cipponi
- Abstract
PDF file - 797K, Positive and Negative controls for the immunohistological detection of the indicated antigens in adjacent cryosections of an inflammatory tonsil
- Published
- 2023
20. Sonidegib improved quality of life in patients with advanced basal cell carcinoma: results from the phase 2 Basal Cell Carcinoma Outcomes with LDE225 Treatment trial through 73 weeks
- Author
-
Michael, Migden, Carmen, Loquai, Caroline, Robert, Jean-François, Baurain, Nicholas, Squittieri, Ramon, Arntz, Jörg, Dierlamm, Brigitte, Dréno, The University of Texas M.D. Anderson Cancer Center [Houston], University Medical Center [Mainz], Institut Gustave Roussy (IGR), Service de dermatologie, Département de médecine oncologique [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Cliniques Universitaires Saint-Luc [Bruxelles], Université Catholique de Louvain = Catholic University of Louvain (UCL), Sun Pharmaceutical Industries, Inc. [Princeton, NJ, USA] (SPI), Sun Pharmaceutical Industries [Hoofddorp, North Holland, Netherlands] (SPI - (Europe) B.V.), Immunology and New Concepts in ImmunoTherapy (INCIT), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Nantes Université - UFR de Médecine et des Techniques Médicales (Nantes Univ - UFR MEDECINE), Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ)-Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ), Service de dermatologie [Nantes], Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes), and Pecqueret, Valérie
- Subjects
[SDV] Life Sciences [q-bio] ,[SDV]Life Sciences [q-bio] ,Dermatology - Abstract
International audience
- Published
- 2022
21. 2022-RA-447-ESGO Efficacy and safety of VB10.16, a therapeutic DNA vaccine specifically targeting antigen-cell presenting cells, in combination with atezolizumab in patients with advanced HPV16-positive cervical cancer: results from a pre-planned interim analysis
- Author
-
Peter Hillemanns, Kristina Lindemann, Jean-François Baurain, Pawel Blecharz, Karsten Bruins Slot, Karoline Schjetne, Siri Torhaug, and Agnete Fredriksen
- Published
- 2022
22. Recurrent High Grade Serous Ovarian Cancer Management
- Author
-
Mathieu Luyckx, Jean-Luc Squifflet, Annika M. Bruger, and Jean-François Baurain
- Published
- 2022
23. Noninvasive detection of the endogenous free radical melanin in human skin melanomas using electron paramagnetic resonance (EPR)
- Author
-
Lionel Mignion, Celine M. Desmet, Evelyne Harkemanne, Isabelle Tromme, Nicolas Joudiou, Mohammad Wehbi, Jean-François Baurain, Bernard Gallez, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
Melanins ,Skin Neoplasms ,Free Radicals ,Physiology (medical) ,Electron Spin Resonance Spectroscopy ,Humans ,Prospective Studies ,Biochemistry ,Melanoma ,Nevus - Abstract
We explored the capability of low-frequency Electron Paramagnetic Resonance (EPR) to noninvasively detect melanin (a stable semiquinone free radical) in the human skin. As previous in vitro studies on biopsies suggested that the EPR signal from melanin was different when measured in skin melanomas or benign nevi, we conducted a prospective first-in-man clinical EPR study in patients with skin lesions suspicious of melanoma. EPR spectra were obtained using a spectrometer operating at 1 GHz, with a surface coil placed over the area of interest. Two clinical studies were carried out: 1) healthy volunteers (n = 45) presenting different skin phototypes; 2) patients (n = 88) with skin lesions suspicious of melanoma (n = 100) requiring surgical resection. EPR data obtained before surgery were compared with histopathology results. The method was not sensitive enough to measure differences in melanin content due to changes in skin pigmentation. In patients, 92% of the spectra were analyzable. The EPR signal of melanin was significantly higher (p < 0.0001) in melanoma lesions (n = 26) than that in benign atypical nevi (n = 62). A trend toward a higher signal intensity (though not significant) was observed in high Breslow depth melanomas (a marker of skin invasion) than in low Breslow lesions. To date, no naturally occurring free radicals have been detected by low-frequency EPR systems adapted for clinical studies. Here, we demonstrated for the first time the ability of this technology to detect an endogenous free radical, opening new avenues for evaluating clinical EPR as a potential aid in the diagnosis of pigmented skin lesions.
- Published
- 2022
24. Safety of systemic anti-cancer treatment in oncology patients with non-severe COVID-19: a cohort study
- Author
-
Emmanuel Seront, M. Van den Eynde, Thierry Pieters, Anaïs Scohy, Philippe Collard, C. Dumont, F. Aboubakar Nana, A. van der Elst, R. Galot, Jean Cyr Yombi, Frank Cornelis, Simon Beyaert, Marco Gizzi, Ivan Borbath, F. Derouane, A. van Maanen, Filomena Mazzeo, N. Whenham, N. Honoré, François Duhoux, A. De Cuyper, Jean-François Baurain, Bertrand Filleul, I. Sinapi, J.-P. Machiels, C. van Marcke, UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique, UCL - SSS/IREC/GAEN - Pôle d'Hépato-gastro-entérologie, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, UCL - SSS/IREC/MBLG - Pôle de Microbiologie médicale, UCL - SSS/IREC/PNEU - Pôle de Pneumologie, ORL et Dermatologie, UCL - SSS/IREC/SLUC - Pôle St.-Luc, UCL - (SLuc) Service de pathologie cardiovasculaire, UCL - (SLuc) Service de microbiologie, UCL - (SLuc) Service de médecine interne générale, UCL - (SLuc) Service de gastro-entérologie, UCL - (SLuc) Service de pneumologie, UCL - (SLuc) Unité d'oncologie médicale, and UCL - (SLuc) Centre du cancer
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Health Personnel ,Population ,Cancer Care Facilities ,law.invention ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Belgium ,Surgical oncology ,law ,Risk Factors ,Internal medicine ,Neoplasms ,Ambulatory ,Genetics ,medicine ,Ambulatory Care ,Humans ,030212 general & internal medicine ,Adverse effect ,education ,RC254-282 ,Aged ,education.field_of_study ,Performance status ,business.industry ,SARS-CoV-2 ,Non-severe COVID-19 ,COVID-19 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Common Terminology Criteria for Adverse Events ,Middle Aged ,medicine.disease ,Systemic anti-cancer treatment ,Intensive care unit ,Oncology ,030220 oncology & carcinogenesis ,Female ,Safety ,business ,Febrile neutropenia ,Cohort study ,Research Article - Abstract
BackgroundThe viral pandemic coronavirus disease 2019 (COVID-19) has disrupted cancer patient management around the world. Most reported data relate to incidence, risk factors, and outcome of severe COVID-19. The safety of systemic anti-cancer therapy in oncology patients with non-severe COVID-19 is an important matter in daily practice.MethodsONCOSARS-1 was a single-center, academic observational study. Adult patients with solid tumors treated in the oncology day unit with systemic anti-cancer therapy during the initial phase of the COVID-19 pandemic in Belgium were prospectively included. All patients (n = 363) underwent severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) serological testing after the first peak of the pandemic in Belgium. Additionally, 141 of these patients also had a SARS-CoV-2 RT-PCR test during the pandemic. The main objective was to retrospectively determine the safety of systemic cancer treatment, measured by the rate of adverse events according to the Common Terminology Criteria for Adverse Events, in SARS-CoV-2-positive patients compared with SARS-CoV-2-negative patients.ResultsTwenty-two (6%) of the 363 eligible patients were positive for SARS-CoV-2 by RT-PCR and/or serology. Of these, three required transient oxygen supplementation, but none required admission to the intensive care unit. Hematotoxicity was the only adverse event more frequently observed in SARS-CoV-2 -positive patients than in SARS-CoV-2-negative patients: 73% vs 35% (P P = 0.009) even after adjusting for performance status and type of systemic treatment. Hematological adverse events led to more treatment delays for the SARS-CoV-2-positive group: 55% vs 20% (P ConclusionSystemic anti-cancer therapy appeared safe in ambulatory oncology patients treated during the COVID-19 pandemic. There were, however, more treatment delays in the SARS-CoV-2-positive population, mainly due to a higher rate of hematological adverse events.
- Published
- 2021
25. Primary analysis of Phase 2 results for cemiplimab in patients with metastatic basal cell carcinoma who progressed on or were intolerant to hedgehog inhibitors
- Author
-
Karl D Lewis, Ketty Peris, Aleksandar Sekulic, Alexander J Stratigos, Lara Dunn, Zeynep Eroglu, Anne Lynn S Chang, Michael R Migden, Suk-Young Yoo, Kosalai Mohan, Ebony Coates, Emmanuel Okoye, Jean-François Baurain, Oliver Bechter, Axel Hauscild, Marcus O Butler, Leonel Hernandez-Aya, Lisa Licitra, Rogerio I Neves, Emily S Ruiz, Frank Seebach, Israel Lowy, Priscila Goncalves, and Matthew G Fury
- Subjects
Dermatology - Published
- 2023
26. Abstract P3-09-19: Preliminary safety data of intrahepatic talimogene laherparepvec and intravenous atezolizumab in patients with triple negative breast cancer
- Author
-
Steven S. Raman, Edward Cha, Jean-François Baurain, Arlene Chan, Miguel Martín, Emily Chan, Kevin Kalinsky, J. R. Hecht, Chunxu Liu, and Federico Longo-Munoz
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,business.industry ,Cancer ,Pembrolizumab ,medicine.disease ,Breast cancer ,Atezolizumab ,Internal medicine ,medicine ,Chills ,medicine.symptom ,business ,Talimogene laherparepvec ,Triple-negative breast cancer - Abstract
Background Talimogene laherparepvec (T-VEC) is a genetically modified, oncolytic herpes simplex virus type 1 designed to selectively replicate within tumors and to produce granulocyte-macrophage colony-stimulating factor to enhance systemic antitumor immune responses. The safety and efficacy of T-VEC in treatment of advanced melanoma has been demonstrated as monotherapy and in combination with the checkpoint inhibitors (CPI) pembrolizumab and ipilimumab. Additionally, T-VEC monotherapy has demonstrated tolerable safety for intrahepatic injection (Hecht GICS 2018). Atezolizumab (atezo) is a humanized monoclonal antibody that promotes antitumor immunity by targeting programmed cell death ligand-1 (PD-L1) and is approved for metastatic triple negative breast cancer (mTNBC). We hypothesize that T-VEC in combination with a CPI may be effective in mTNBC and CRC in addition to melanoma. This phase 1b, multicenter study (clinicaltrials.gov identifier: NCT03256344) evaluates the safety of intrahepatic injection of T-VEC in combination with intravenous (IV) atezo in patients (pts) with mTNBC or colorectal cancer (CRC) with liver metastases (LMs). Here we report early safety data in the TNBC cohort. Methods Key eligibility criteria included age ≥ 18 years, confirmed diagnosis of TNBC or CRC with LMs, ECOG PS 0/1, adequate organ function, disease progression during or after ≥ 1 prior standard-of-care systemic therapy for metastatic disease, and ≥ 1 measurable LM suitable for injection. T-VEC (≤ 4mL) was injected by image-guided intralesional injection: 106 PFU/mL day 1, 108 PFU/mL every 21 days thereafter; atezo 1,200 mg IV was given on day 1 and every 21 days thereafter. The primary objective of this study is to evaluate incidence of dose limiting toxicity (DLT) for safety of T-VEC injection into LMs in combination with IV atezo, by tumor type. The DLT analysis set included DLT-evaluable pts who were on treatment for at least 6 weeks from the initial dose of study treatment and received at least 2 doses of T-VEC and 2 doses of atezo in combination or have a DLT during the DLT evaluation period. Key secondary objectives include objective response rate, durable response rate, progression-free survival, and overall survival of the combination therapy, by tumor type. Results Thirty-two pts were enrolled in two parallel cohorts (8 TNBC, 24 CRC). Of the 8 TNBC pts, 4 were evaluable for DLT at the time of this analysis. Of the 4 pts that were not DLT-evaluable, 1 never received study drug, 1 received atezo monotherapy, and 2 received intrahepatic T-VEC and IV atezo but stopped study drugs for unconfirmed progression before the DLT evaluation period was completed. No DLTs were reported in the TNBC cohort. In the 7 TNBC pts who received at least one dose of the study drug, the most common treatment-emergent adverse events (TEAEs) in terms of the subject incidence were pyrexia (71.4%), chills (42.9%), and rash (42.9%). In the 7 TNBC pts who received at least one dose of study drug, T-VEC related AEs in more than one pt were pyrexia (n=3) and chills (n=2). Atezo related AEs in more than one pt were pyrexia (n=5), chills and rash (n=3 each), and fatigue, arthralgia, pain, and pruritus (n=2 each). Study drug-related serious adverse events (SAEs) occurred in 3 (42.9%) pts, including Grade (G) 3 hypersensitivity related to atezo, G3 orthostatic hypotension related to T-VEC and atezo, and G1 pyrexia related to T-VEC and atezo. Procedure-related SAEs occurred in 1 (14.3%) pt, of G3 hepatic hematoma and G3 abdominal infection, but these were not related to the study drugs. One confirmed partial response in the TNBC cohort has been seen thus far. Conclusion T-VEC intrahepatic injection in combination with IV atezo at standard doses has thus far been demonstrated as feasible and tolerable with no DLTs observed in the TNBC cohort to date. Citation Format: Joel Randolph Hecht, Arlene Chan, Jean-Francois Baurain, Miguel Martin, Federico Longo-Munoz, Kevin Kalinsky, Steven Raman, Chunxu Liu, Edward Cha, Emily Chan. Preliminary safety data of intrahepatic talimogene laherparepvec and intravenous atezolizumab in patients with triple negative breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P3-09-19.
- Published
- 2020
27. Combined HP
- Author
-
Chantale, Farah, Marie-Aline, Neveu, Caner, Yelek, Caroline, Bouzin, Bernard, Gallez, Jean-François, Baurain, Lionel, Mignion, and Bénédicte F, Jordan
- Abstract
A vast majority of BRAF V600E mutated melanoma patients will develop resistance to combined BRAF/MEK inhibition after initial clinical response. Resistance to targeted therapy is described to be accompanied by specific metabolic changes in melanoma. The aim of this work was to evaluate metabolic imaging using
- Published
- 2022
28. When a metastatic breast cancer is mimicking a pancreatic cancer: case report and review of the literature
- Author
-
Etienne Danse, Jean Cyr Yombi, Mina Komuta, Halil Yildiz, Jean-François Baurain, Françoise Derouane, UCL - SSS/IREC/GAEN - Pôle d'Hépato-gastro-entérologie, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, UCL - SSS/IREC/IMAG - Pôle d'imagerie médicale, UCL - SSS/IREC/SLUC - Pôle St.-Luc, UCL - (SLuc) Service d'anatomie pathologique, UCL - (SLuc) Service de médecine interne générale, UCL - (SLuc) Unité d'oncologie médicale, UCL - (SLuc) Service de radiologie, and UCL - (SLuc) Centre du cancer
- Subjects
Oncology ,medicine.medical_specialty ,Paclitaxel ,Biopsy ,Jaundice ,Breast Neoplasms ,ductal carcinoma ,Antibodies, Monoclonal, Humanized ,Diagnosis, Differential ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Pancreatic cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Weight Loss ,medicine ,Humans ,Retroperitoneal Space ,030212 general & internal medicine ,Stage (cooking) ,skin and connective tissue diseases ,Ultrasonography ,Pancreatic metastases ,Cholangiopancreatography, Endoscopic Retrograde ,Common Bile Duct ,business.industry ,Carcinoma, Ductal, Breast ,Liver Neoplasms ,General Medicine ,Middle Aged ,Trastuzumab ,Ductal carcinoma ,medicine.disease ,Metastatic breast cancer ,Pancreatic Neoplasms ,Ductal Breast Carcinoma ,030220 oncology & carcinogenesis ,Female ,Lymph Nodes ,medicine.symptom ,Presentation (obstetrics) ,Tomography, X-Ray Computed ,business - Abstract
We report the case of a 51 year-old female who complained of jaundice and weight loss. At the time of presentation, she had been in remission from a stage 2a ductal breast carcinoma for 58 months. The clinical presentation was suggestive of a primary pancreas cancer with liver metastases and retroperitoneal lymph nodes. We performed liver and pancreas biopsies that demonstrate a relapse of her old breast carcinoma with positive hormone receptors and HER2 positive. Conservative treatment by chemotherapy was given with Paclitaxel - Trastuzumab and Pertuzumab. Pancreatic metastases are uncommon. Furthermore, pancreatic metastases from breast cancer are very rare. We performed a review of the literature and found 48 cases of pancreatic metastases from breast cancer. We would like to highlight by this case that when a pancreatic lesion appears, in patients with a past history of cancer, physicians must not forget the possibility of metastases from primary tumor even if the initial stage, of the tumor, is low. However the diagnosis is not always easy. If liver and pancreatic lesions occur simultaneously, the clinical presentation can mimic metastatic primary pancreatic cancer. Therefore performing biopsy is highly recommended for making the correct diagnosis and also for the staging of the disease and the choice of the best treatment according to immunohistochemical analysis.
- Published
- 2019
29. Abstract CT165: Primary analysis of phase 2 results for cemiplimab in patients with metastatic basal cell carcinoma who progressed on or were intolerant to hedgehog inhibitors
- Author
-
Karl Lewis, Ketty Peris, Aleksander Sekulic, Alexander J. Stratigos, Lara Dunn, Zeynep Eroglu, Anne Lynn S. Chang, Michael R. Migden, Suk-Young Yoo, Kosalai Mohan, Ebony Coates, Emmanuel Okoye, Timothy Bowler, Jean-François Baurain, Oliver Bechter, Axel Hauschild, Marcus O. Butler, Leonel Hernandez-Aya, Lisa Licitra, Rogerio I. Neves, Emily S. Ruiz, Frank Seebach, David M. Weinreich, George D. Yancopoulos, Israel Lowy, Priscila Goncalves, and Matthew G. Fury
- Subjects
Cancer Research ,Oncology - Abstract
Background: Cemiplimab is the first treatment approved in the US (as cemiplimab-rwlc) for patients with metastatic basal cell carcinoma (mBCC) or locally advanced disease (laBCC) after hedgehog inhibitor (HHI) treatment or for whom an HHI is not appropriate. Cemiplimab provided substantial clinical benefit and an acceptable safety profile in patients with laBCC who discontinued HHI therapy due to progressive disease (PD), intolerance, or no better than stable disease (SD) after 9 months (NCT03132636). Here, we present the primary analysis of the mBCC cohort. Methods: Patients with mBCC (nodal or distant) post-HHI treatment received cemiplimab 350 mg intravenously every 3 weeks for up to 93 weeks or until PD. The primary endpoint was objective response rate (ORR) by independent central review (ICR). Key secondary endpoints included safety and tolerability, ORR per investigator (INV) assessment, duration of response (DOR), progression-free survival (PFS), overall survival (OS), and complete response (CR) rate (data cutoff date: May 20, 2021). Results: Fifty-four patients were enrolled (70.4% male; median age 63.5 years [range, 38−90]; Eastern Cooperative Oncology Group performance status 0 [66.7%] and 1 [33.3%]). Median duration of follow-up was 8.4 months (range 1.5-36.2). ORR per ICR was 24.1% (95% confidence interval [CI], 13.5-37.6); with 1 CR and 12 partial responses (PRs). ORR per INV was 25.9% (95% CI, 15.0-39.7), with 2 CRs and 12 PRs. Among responders, median time to response per ICR was 4.0 months (range, 2.0−10.5). Estimated median DOR per ICR was 16.7 months (95% CI, 9.8-not evaluable). Disease control rate was 63.0% (95% CI, 48.7-75.7) per ICR and 70.4% (95% CI, 56.4-82.0) per INV. Median OS was not reached. Median PFS per ICR was 8.3 months (95% CI, 4.2-15.9). The most common treatment-related adverse events were fatigue (37.0%), diarrhea (14.8%), pruritus (13.0%), hyperthyroidism (9.3%), and arthralgia (9.3%) as well as hypothyroidism, asthenia, constipation, and maculo-papular rash (7.4% each). There were no treatment-related deaths. Conclusions: Cemiplimab provided clinically meaningful antitumor activity, including durable responses, and an acceptable safety profile in patients with mBCC who had progressed on or were intolerant to HHI therapy. Citation Format: Karl Lewis, Ketty Peris, Aleksander Sekulic, Alexander J. Stratigos, Lara Dunn, Zeynep Eroglu, Anne Lynn S. Chang, Michael R. Migden, Suk-Young Yoo, Kosalai Mohan, Ebony Coates, Emmanuel Okoye, Timothy Bowler, Jean-François Baurain, Oliver Bechter, Axel Hauschild, Marcus O. Butler, Leonel Hernandez-Aya, Lisa Licitra, Rogerio I. Neves, Emily S. Ruiz, Frank Seebach, David M. Weinreich, George D. Yancopoulos, Israel Lowy, Priscila Goncalves, Matthew G. Fury. Primary analysis of phase 2 results for cemiplimab in patients with metastatic basal cell carcinoma who progressed on or were intolerant to hedgehog inhibitors [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 CT165.
- Published
- 2022
30. 347 Efficacy and safety of niraparib in older patients (PTS) with advanced ovarian cancer (OC): results from the prima/ENGOT-OV26/GOG-3012 trial
- Author
-
Florian Heitz, Hanna Dahlstrand, Whitney Graybill, Bhavana Pothuri, Bradley J. Monk, Jean-François Baurain, Thibault de La Motte Rouge, Li Yong, Antonio González-Martín, and Colleen McCormick
- Subjects
Oncology ,medicine.medical_specialty ,Advanced ovarian cancer ,business.industry ,Newly diagnosed ,Regimen ,Older patients ,Age groups ,Precision oncology ,Internal medicine ,Partial response ,medicine ,business ,Protocol Amendment - Abstract
Introduction/Background The PRIMA/ENGOT-OV26/GOG-3012 (PRIMA) trial showed that niraparib significantly improves progression-free survival (PFS) in pts with newly diagnosed advanced OC that responded to first-line platinum-based chemotherapy (CT) (hazard ratio [HR] 0.62; 95% CI 0.50–0.76). Here we discuss the impact of age on efficacy and safety of niraparib. Methodology This double-blind, placebo (PBO)-controlled phase 3 trial evaluated niraparib in pts with newly diagnosed, advanced, high-grade serous or endometrioid ovarian, primary peritoneal, or fallopian tube cancer with a complete or partial response to first-line platinum-based CT. Pts were randomised 2:1 to receive either a fixed starting dose (FSD) of 300 mg niraparib or PBO QD. A protocol amendment introduced an individualised starting dose (ISD): 200 mg QD in pts with bodyweight Results Of 733 enrolled pts, 444 were Conclusion Niraparib efficacy, safety, and QOL were similar in compared age groups. Implementation of an ISD regimen improved rates of grade ≥3 thrombocytopenia events in older pts. Disclosures Funding: GlaxoSmithKline NCT number: NCT02655016 Encore statement: This data is presented on behalf of the original authors with their permission. Presented at the European Society for Medical Oncology (ESMO) Annual Meeting, September 19–21, 2020, Virtual. Dr. Dahlstrand reports personal fees from AstraZeneca and Roche. Dr. Pothuri reports grants, personal fees and non-financial support from GSK; Advisory Board fees from AstraZeneca and Clovis Oncology. Dr. Graybill reports personal fees from GSK. Dr. McCormick has nothing to disclose. Dr. de La Motte Rouge reports personal fees and non-financial support from ASTRAZENECA, MSD, and Roche; personal fees from Clovis Oncology and GlaxoSmithKline; and grants, personal fees, and non-financial support from Pfizer. Dr. Heitz reports non-financial support from NewOncology; Personal fees from Roche, AstraZeneca, Clovis, Tesaro, and PharmaMar. Dr. Monk reports consulting and advisory role at Merck, GSK, Roche/Genentech, AstraZeneca, Advaxiz, Cerulean Pharma, Amgen, Immunogen, NuCana BioMed, Clovis Oncology, Pfizer, Mateon Therapeutics, Precision Oncology, Perthera, Abbvie, Myriad Pharmaceuticals, Incyte, VBL Therapeutics, Takeda, Samumed, Oncomed, OncoSec, ChemoID, Geistlich Pharma, Eisai and Chemocare; Speakers’ bureau at Roche/Genentech, AstraZeneca, Janssen, Clovis Oncology and GSK; Honoraria from Merck, GSK, Roche/Genentech, AstraZeneca, Advaxis, Immunogen, NuCana BioMed, Clovis Oncology, Pfizer, Mateon Therapeutics, Precision Oncology, Pethera, Abbvie, Myriad Pharmaceuticals, Incyte, Janssen, Amgen, Genmab, Samumed, Takeda, VBL Therapeutics, Puma Biotechnology, Immunomedics, Conjupro Biotherapeutics, Agenus, OncoQuest, ChemoID, Geistlich Pharma, Eisai and Chemocare; and Research funding from Novartis, Amgen, Genentech, Lilly, Janssen, Array BioPharma, GSK, Morphotek, Pfizer, Advaxis, AstraZeneca, Immunogen, Regeneron, and Nucana. Dr. Gonzalez-Martin reports personal fees and non-financial support from AstraZeneca; Grant and personal fees from GSK, Clovis Oncology, Roche Holding AG, Merck & Co., Inc., Genmab, INMUNOGEN, Pharma Mar, S.A., and Oncoinvent AS. Drs Li and Gupta are employees of GlaxoSmithKline.
- Published
- 2020
31. 388 Efficacy of individualised starting dose (ISD) and fixed starting dose (FSD) of niraparib per investigator assessment (IA) in newly diagnosed advanced ovarian cancer (OC) patients
- Author
-
Divya Gupta, Ulla Peen, Emily Prendergast, Antonio González-Martin, Whitney Graybill, Bradley J. Monk, Jean-François Baurain, Elena Ioana Braicu, David M. O’Malley, and Philippe Follana
- Subjects
Oncology ,Advanced ovarian cancer ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Newly diagnosed ,business - Published
- 2020
32. Safety of systemic anti-cancer treatment in oncology patients with non-severe COVID-19: a prospective cohort study
- Author
-
Bertrand Filleul, N. Whenham, Frank Aboubakar Nana, Philippe Collard, Athénaïs van der Elst, Marco Gizzi, Marc Van den Eynde, Cédric van Marcke, Rachel Galot, Jean Cyr Yombi, Jean-François Baurain, Aline Van Maanen, François Duhoux, Emmanuel Seront, N. Honoré, Jean-Pascal Machiels, Caroline Dumont, I. Sinapi, Simon Beyaert, Françoise Derouane, Thierry Pieters, Anaïs Scohy, Ivan Borbath, Filomena Mazzeo, Astrid De Cuyper, and Frank Cornelis
- Subjects
medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Internal medicine ,medicine ,Oncology patients ,Prospective cohort study ,business ,Cancer treatment - Abstract
Background The viral pandemic coronavirus disease 2019 (COVID-19) has disrupted cancer patient management around the world. Most reported data relate to incidence, risk factors, and outcome of severe COVID-19. The safety of systemic anti-cancer therapy in oncology patients with non-severe COVID-19 is unknown.Methods ONCOSARS-1 was a prospective, single-center, academic observational study. Adult patients with solid tumors treated in the oncology day unit with systemic anti-cancer therapy during the initial phase of the COVID-19 pandemic in Belgium were included. All patients (n=363) underwent severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) serological testing after the peak of the pandemic in Belgium. Additionally, 141 of these patients also had a SARS-CoV-2 RT-PCR test during the pandemic. The main objective was to determine the safety of systemic cancer treatment, measured by the rate of adverse events according to the Common Terminology Criteria for Adverse Events, in SARS-CoV-2-positive patients compared with SARS-CoV-2-negative patients.Results Twenty-two (6%) of the 363 eligible patients were positive for SARS-CoV-2 by RT-PCR and/or serology. Of these, three required transient oxygen supplementation, but none required admission to the intensive care unit. Hematotoxicity was the only adverse event more frequently observed in SARS-CoV-2 -positive patients than in SARS-CoV-2-negative patients: 73% vs 35% (PP=0.009) even after adjusting for performance status and type of systemic treatment. Hematological adverse events led to more treatment delays for the SARS-CoV-2-positive group: 55% vs 20% (PConclusion Systemic anti-cancer therapy appeared safe in ambulatory oncology patients treated during the COVID-19 pandemic. There were, however, more treatment delays in the SARS-CoV-2-positive population, mainly due to a higher rate of hematological adverse events.
- Published
- 2020
33. 428 Interim analysis of Phase 2 results for cemiplimab in patients with metastatic basal cell carcinoma (mBCC) who progressed on or are intolerant to hedgehog inhibitors (HHIs)
- Author
-
Leonel Hernandez-Aya, Michael R. Migden, Aleksandar Sekulic, Oliver Bechter, Rogerio I. Neves, Kosalai Kal Mohan, Timothy Bowler, Axel Hauschild, Zeynep Eroglu, Ketty Peris, David M. Weinreich, Israel Lowy, Lisa Licitra, Siyu Li, Karl D. Lewis, Jean-François Baurain, Frank Seebach, Marcus O. Butler, Lara Dunn, George D. Yancopoulos, Emily S. Ruiz, Ebony Coates, Anne Lynn S. Chang, Emmanuel Okoye, Matthew G. Fury, and Alexander J. Stratigos
- Subjects
medicine.medical_specialty ,business.industry ,Vismodegib ,Institutional review board ,Interim analysis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Sonidegib ,chemistry.chemical_compound ,Tolerability ,chemistry ,Internal medicine ,Cohort ,medicine ,Clinical endpoint ,Adverse effect ,business ,medicine.drug - Abstract
Background HHIs, vismodegib and sonidegib, are approved for treatment of patients with mBCC or locally advanced BCC who are not candidates for surgery or radiation. There is no approved option for patients who progress on or are intolerant to HHIs. Cemiplimab is an anti-programmed cell death-1 monoclonal antibody approved for treatment of patients with metastatic cutaneous squamous cell carcinoma (CSCC) or locally advanced CSCC who are not candidates for curative surgery or curative radiation. Here we present the prespecified interim analysis of the mBCC cohort from the pivotal Phase 2, non-randomized, multi-center study of cemiplimab in patients with advanced BCC who discontinued HHI therapy due to disease progression, intolerance, or no better than stable disease after 9 months (NCT03132636). Methods Patients with mBCC (nodal and/or distant) received cemiplimab 350 mg intravenously every 3 weeks; interim analysis included patients with the opportunity to be followed for approximately 57 weeks. The primary endpoint was objective response rate (ORR) per independent central review (ICR). Secondary objectives included assessment of safety and tolerability, estimation of duration of response (DOR), progression-free survival (PFS), and overall survival (OS). Results In this interim efficacy analysis of 28 patients, 82.1% were males and median age was 65.5 years (range 38−90). Six patients had a partial response, per ICR, for an ORR of 21.4% (95% CI, 8.3, 41.0). ORR per investigator assessment was 28.6% (95% CI, 13.2, 48.7). Among responders, observed DOR was 9−23 months. Median time to response per ICR was 3.2 months (range, 2.1−10.5). Median Kaplan–Meier (KM) estimation of PFS was 8.3 months. Median DOR had not been reached and median KM estimation of OS was 25.7 months. All six responses had observed durations of at least 8 months. The disease control rate was 67.9% (95% CI, 47.6, 84.1).The most common treatment emergent adverse events (TEAEs) regardless of attribution were fatigue (50.0%), diarrhea (35.7%), pruritus (25.0%), and constipation (25.0%). Hypertension (n=2) was the only Grade ≥3 TEAE regardless of attribution occurring in ≥2 patients. TEAEs leading to death occurred in one (3.6%) patient who died from staphylococcal pneumonia, considered unrelated to study treatment. Conclusions This interim analysis demonstrates that cemiplimab is the first agent to provide clinically meaningful anti-tumor activity, including durable responses, in patients with mBCC after progression or intolerance on HHI therapy. Acknowledgements Editorial acknowledgment: Medical writing support was provided by Cindi Hoover, PhD of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc. and Sanofi. Ethics Approval The study protocols and all amendments were approved by the institutional review board at each participating study site. The study was conducted in accordance with the principles of the Declaration of Helsinki and with Good Clinical Practice guidelines as defined by the International Conference on Harmonization. All patients provided written informed consent before enrollment.
- Published
- 2020
34. 391 A first-in-human study of intratumoral SAR441000, an mRNA mixture encoding IL-12sc, interferon alpha2b, GM-CSF and IL-15sushi as monotherapy and in combination with cemiplimab in advanced solid tumors
- Author
-
Oliver Bechter, Evelyna Derhovanessian, Rahul Marpadga, Esteban-Rodrigo Imedio, Ugur Sahin, Nicolas Acquavella, Carmen Loquai, Christophe Massard, Jochen Utikal, Marie-Laure Ozoux, and Jean-François Baurain
- Subjects
Combination therapy ,business.industry ,Melanoma ,medicine.medical_treatment ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Proinflammatory cytokine ,Cytokine ,Immune system ,Interferon ,medicine ,Cancer research ,Interferon gamma ,Sarcoma ,business ,medicine.drug - Abstract
Background mRNA-based-drugs can be applied for cancer immunotherapy.1 SAR441000 is a novel saline-formulated mixture of four mRNAs encoding interleukin-12 single chain, interferon alpha-2b, granulocyte-macrophage colony-stimulating factor, and interleukin-15 sushi that we have identified as mediators of tumor regression across different murine tumor models. Local intratumoral administration of SAR441000 in immunocompetent mice, mediates successful antitumor immunity leading to tumor eradication. Effective antitumor activity of these cytokines involved multiple immune cell populations and was accompanied by intratumoral interferon gamma induction, systemic antigen-specific T-cell expansion, increased granzyme B+ T-cell infiltration, and formation of immune memory. Antitumor activity extended beyond the treated lesions and inhibited growth of non-injected distant tumors. Combining the mRNAs with checkpoint inhibitors enhanced antitumor responses in both injected and non-injected tumors, improving survival and tumor regression in mice. Based on these preclinical observations a clinical study was initiated. Methods In a phase 1 dose escalation study, patients with advanced solid tumors were treated with weekly intratumoral administration of SAR441000 monotherapy and in combination with fixed dose of cemiplimab 350 mg. Plasma samples for cytokine analysis and tumor biopsies were collected at baseline and throughout the study to characterize the PK/PD profile of SAR441000, immune cell tumor infiltration by immunohistochemistry and the presence of corresponding tumor proinflammatory signatures by RNA sequencing. Results As of July 2020, 17 patients received SAR441000 monotherapy (melanoma 7, breast 4, sarcoma 2, Cutaneous Squamous Cell 2, Basal Cell 1, and Merkel Cell 1) at dose levels 1 through 7. Six patients received SAR441000 in combination therapy (melanoma 3, breast 3) at dose levels 4 and 5. No patient experienced a Dose Limiting Toxicity. No grade 3, 4 or 5 adverse events related to study treatment were reported. Adverse events related to study treatment in two or more subjects in both treatment groups combined were nonserious grade 1 or 2 fatigue (43%;10/23), vomiting (17%; 4/23), nausea (13%;3/23); local injection site reaction (11.7%, 2/23); and chills, diarrhea, and rash were reported as 9% (2/23), respectively (table 1 and 2). In some patients, increases in plasma IP10 and IFN gamma and CD8+ T cell infiltration in tumor biopsies were observed. Conclusions SAR441000 administered as monotherapy and in combination with cemiplimab was generally well tolerated. An immunomodulatory effect is suggested by downstream effector cytokines and T cell infiltration. These data support further clinical evaluation of SAR441000. Ethics Approval The study was approved by each participating Institution’s Ethics or Institutional Review Board(s). Reference Sahin U, Kariko K, Tureci O. mRNA-based therapeutics-developing a new class of drugs. Nat. Rev. Drug Discov 2014;13:759–780.
- Published
- 2020
35. Recommendations for skin cancer consultation and surgery during COVID‐19 pandemic
- Author
-
Arjen Nikkels, M. Garmyn, François Sales, A. Van Laethem, Marguerite Stas, Vibeke Kruse, Lieve Brochez, V. Del Marmol, and Jean-François Baurain
- Subjects
Dermatologie ,medicine.medical_specialty ,2019-20 coronavirus outbreak ,Skin Neoplasms ,Coronavirus disease 2019 (COVID-19) ,business.industry ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,MEDLINE ,COVID-19 ,Dermatology ,medicine.disease ,Infectious Diseases ,Pandemic ,medicine ,Commentary ,Humans ,Skin cancer ,Intensive care medicine ,business ,Pathologie maladies infectieuses ,Pandemics ,Referral and Consultation - Abstract
SCOPUS: no.j, info:eu-repo/semantics/published
- Published
- 2020
36. Metabolic imaging using hyperpolarized 13 C‐pyruvate to assess sensitivity to the B‐Raf inhibitor vemurafenib in melanoma cells and xenografts
- Author
-
Florian Gourgue, Jean-François Baurain, Céline A Schoonjans, Caroline Bouzin, Nicolas Joudiou, Bénédicte F. Jordan, Stefania Acciardo, Bernard Gallez, Lionel Mignion, Estelle Lacomblez, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
0301 basic medicine ,Chemistry ,Melanoma ,Cell Biology ,medicine.disease ,In vitro ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Metabolic Marker ,Immunohistochemistry ,Molecular Medicine ,Glycolysis ,Vemurafenib ,Ex vivo ,medicine.drug - Abstract
Nearly all melanoma patients with a BRAF-activating mutation will develop resistance after an initial clinical benefit from BRAF inhibition (BRAFi). The aim of this work is to evaluate whether metabolic imaging using hyperpolarized (HP) 13 C pyruvate can serve as a metabolic marker of early response to BRAFi in melanoma, by exploiting the metabolic effects of BRAFi. Mice bearing human melanoma xenografts were treated with the BRAFi vemurafenib or vehicle. In vivo HP 13 C magnetic resonance spectroscopy was performed at baseline and 24 hours after treatment to evaluate changes in pyruvate-to-lactate conversion. Oxygen partial pressure was measured via electron paramagnetic resonance oximetry. Ex vivo qRT-PCR, immunohistochemistry and WB analysis were performed on tumour samples collected at the same time-points selected for in vivo experiments. Similar approaches were applied to evaluate the effect of BRAFi on sensitive and resistant melanoma cells in vitro, excluding the role of tumour microenvironment. BRAF inhibition induced a significant increase in the HP pyruvate-to-lactate conversion in vivo, followed by a reduction of hypoxia. Conversely, the conversion was inhibited in vitro, which was consistent with BRAFi-mediated impairment of glycolysis. The paradoxical increase of pyruvate-to-lactate conversion in vivo suggests that such conversion is highly influenced by the tumour microenvironment.
- Published
- 2020
37. Niraparib exposure-response relationship in patients (pts) with newly diagnosed advanced ovarian cancer (AOC)
- Author
-
Peter G. Rose, Ignacio A. Romero, David M. O'Malley, Oi Wah S. Yap, Bente Lund, Bradley J. Monk, Antonio Gonzalez Martin, Cristina Maria Churruca, Amnon Amit, Elena Ioana Braicu, Emily Prendergast, Whitney Graybill, Hannelore Denys, Michel Fabbro, Zhi-Yi Zhang, Carmela Pisano, Ashley Milton, Sharad A. Ghamande, Paula Calvert, and Jean-François Baurain
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Advanced ovarian cancer ,business.industry ,medicine.medical_treatment ,Newly diagnosed ,Partial response ,Internal medicine ,medicine ,In patient ,business ,Exposure response - Abstract
6051 Background: Niraparib improves progression-free survival (PFS) in pts with newly diagnosed AOC after complete or partial response to first-line, platinum-based chemotherapy. In the PRIMA/ENGOT-OV26/GOG-3012 (PRIMA) trial, pts were treated with a fixed starting dose (FSD) of 300 mg QD until a protocol amendment introduced the individualized starting dose (ISD) regimen: 200 mg QD for pts with baseline bodyweight (BW) < 77 kg and/or platelet count (PC) < 150,000/µL, or 300 mg QD for pts with baseline BW ≥77 kg and PC ≥150,000/µL. Here, we developed a population pharmacokinetic (PopPK) model for niraparib and evaluated exposure-response relationships for pts receiving niraparib using safety and efficacy data from PRIMA. Methods: The PopPK model for niraparib was developed based on 7418 plasma samples from 1442 pts from 4 studies: PN001, NOVA, QUADRA, and PRIMA. PRIMA PK samples were collected on cycle 1, day 1 (C1D1), C2D1 pre-dose and 2 h post-dose, C4D1, and C8D1 pre-dose (or EOT if patient discontinued before C8D1). The relationship between PopPK model-based prospective exposure (average concentration [ Cave] until progression/death) and efficacy (PFS) were evaluated in pts receiving niraparib in both the homologous-recombination deficient (HRd) and overall population. The relationship between model-predicted exposure metrics and incidence of clinically relevant adverse events (AEs) was analyzed using univariate logistic regression in pts receiving niraparib. Results: Of 484 pts receiving niraparib in PRIMA, 480 had PK data and were included in the efficacy and safety analysis. The safety exposure-response showed significant associations ( p≤0.0128) between increasing niraparib exposure and increasing probability of experiencing any-grade and grade ≥3 AEs, except grade ≥3 hypertension. The incidence of AEs, including thrombocytopenia, was lower in pts who received a 200-mg ISD. Efficacy was not compromised in these pts. Conclusions: Niraparib exposure was associated with increased risk of select AEs. However, the ISD regimen decreased AE risk without compromising efficacy. Clinical trial information: NCT02655016.
- Published
- 2020
38. Imaging markers of response to combined BRAF and MEK inhibition in BRAF mutated vemurafenib-sensitive and resistant melanomas
- Author
-
Bénédicte F. Jordan, Bernard Gallez, Jean-François Baurain, Lionel Mignion, Caroline Bouzin, Nicolas Joudiou, Stefania Acciardo, UCL - SSS/LDRI - Louvain Drug Research Institute, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, and UCL - (SLuc) Unité d'oncologie médicale
- Subjects
BRAF/MEK inhibitors ,Combination therapy ,choline spectroscopy ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,melanoma ,medicine ,Distribution (pharmacology) ,Choline ,Vemurafenib ,Trametinib ,tumor response ,business.industry ,Melanoma ,medicine.disease ,diffusion-weighted MRI ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Cancer research ,Immunohistochemistry ,business ,Research Paper ,medicine.drug - Abstract
A majority of patients with a V600x melanoma respond quickly to BRAF/MEK inhibition (BRAFi/MEKi) and have an obvious clinical benefit. Nearly all the patients after this initial phase will develop resistance. Therefore, non-invasive early markers of response/non-response are needed in order to identify those patients who, due to intrinsic or acquired resistance, do not respond to treatment and would be eligible for alternative treatments. The aim of this study was to investigate the value of magnetic resonance spectroscopy (1H-MRS) of choline and diffusion-weighted magnetic resonance imaging (DW-MRI) as early markers of response to BRAF inhibition (BRAFi) with vemurafenib alone or in combination with MEK inhibition (MEKi) with trametinib, in BRAFi-sensitive and BRAFi-resistant melanoma xenografts. Tumor response was significantly improved by the combination of BRAFi and MEKi, compared to BRAFi alone, only in sensitive xenografts; thus indicating that vemurafenib-resistant A375R xenografts were cross-resistant to the inhibition of MEK, as confirmed by immunohistochemistry analysis for phosphorylated ERK. In vivo 1H-MRS showed that in sensitive melanoma xenografts, a significant blockage of ERK phosphorylation, but not a decrease in cell proliferation, was required to affect total choline (tCho) levels, thus suggesting that tCho could serve as a pharmacodynamic (PD) marker for agents targeting the MAPK cascade. In addition, early effects of the combination therapy on tumor cellularity could be detected via DW-MRI. In particular, skewness and kurtosis of the apparent diffusion coefficient (ADC) distribution may be useful to detect changes in the diffusional heterogeneity that might not affect the global ADC value.
- Published
- 2018
39. Abstract CT002: Phase 3 randomized trial comparing tebentafusp with investigator's choice in first line metastatic uveal melanoma
- Author
-
Jessica C. Hassel, Marcus O. Butler, Reinhard Dummer, Anthony M. Joshua, John M. Kirkwood, Paul Nathan, Max Schlaak, Richard D. Carvajal, Howard Goodall, Omid Hamid, Marlana Orloff, Piotr Rutkowski, Alexander N. Shoushtari, Shaad Essa Abdullah, Chris Holland, Sebastian Ochsenreither, Jean-François Baurain, Sophie Piperno-Neumann, Ryan J. Sullivan, and Joseph J. Sacco
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Dacarbazine ,Population ,Ipilimumab ,Pembrolizumab ,Interim analysis ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Clinical endpoint ,Progression-free survival ,education ,business ,medicine.drug - Abstract
Background: Metastatic uveal melanoma (mUM) has a poor prognosis with a 1-yr OS rate of 52%. No systemic treatment has proven an OS benefit in randomized trials. Tebentafusp (tebe), a bispecific consisting of an affinity-enhanced T cell receptor (TCR) fused to an anti-CD3 effector that can redirect T cells to target gp100+ cells, has shown promising activity in previously treated mUM pts. Here, we report the primary analysis of overall survival (OS) in the intention-to-treat population (ITT) of a Ph3 trial of tebe vs. investigator's choice (IC) as first line (1L) therapy in pts with mUM [NCT03070392]. Materials and Methods: In this randomized, open-label, Ph3 trial, 1L HLA-A*02:01+ pts with mUM were randomized 2:1 to receive tebe or IC of pembrolizumab, ipilimumab or dacarbazine, stratified by LDH. The primary endpoint was OS, defined as the time from randomization to death from any cause. Dual primary objectives were to evaluate 1) OS in the ITT population by comparing all tebe-randomized pts to all IC-randomized pts; and 2) OS in tebentafusp-treated patients with rash during week 1 versus all IC-treated patients. Secondary endpoints included safety and RECIST-defined overall response rate (ORR), progression free survival (PFS) and disease control rate (DCR). Here we present the OS in the ITT population. The study was unblinded by an independent data monitoring committee at the first pre-specified interim analysis. Investigator-reported radiographic-based endpoints were not mature at the first interim analysis. This analysis was conducted on the first interim analysis (data extracted Nov 2020). Results: 378 pts were randomized to tebe (252) or IC, including pembrolizumab (103), ipilimumab (15) or dacarbazine (7). Tebe significantly prolonged OS compared to IC (HR 0.51; 95% CI 0.36-0.71; PULN and versus pembrolizumab IC. Most common TRAEs were skin-related (gp100+ melanocytes) or cytokine-mediated (T cell activation) and included pyrexia, pruritus, and rash. These AEs decreased in frequency and severity after the first 3-4 doses and were generally manageable with standard interventions. In the tebe arm, the rate of treatment discontinuation due to TRAEs was low ( Conclusions: In 1L treatment of mUM pts, tebe monotherapy significantly improved OS compared to IC; the first investigational therapy to improve OS in pts with mUM. Tebe had a predictable and manageable AE profile with a low rate of related discontinuation. Tebe is the first TCR therapeutic to demonstrate an OS benefit. Citation Format: Sophie Piperno-Neumann, Jessica C. Hassel, Piotr Rutkowski, Jean-Francois Baurain, Marcus O. Butler, Max Schlaak, Ryan J. Sullivan, Sebastian Ochsenreither, Reinhard Dummer, John M. Kirkwood, Anthony M. Joshua, Joseph J. Sacco, Alexander N. Shoushtari, Marlana Orloff, Richard D. Carvajal, Omid Hamid, Shaad E. Abdullah, Chris Holland, Howard Goodall, Paul Nathan. Phase 3 randomized trial comparing tebentafusp with investigator's choice in first line metastatic uveal melanoma [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 CT002.
- Published
- 2021
40. Highly Aggressive Metastatic Melanoma Cells Unable to Maintain Telomere Length
- Author
-
Marie-Sophie Dheur, Nicolas van Baren, Jean-François Baurain, Nikenza Viceconte, Anabelle Decottignies, Christophe Pierreux, Eva Majerova, and UCL - SSS/DDUV - Institut de Duve
- Subjects
Adult ,Male ,0301 basic medicine ,Telomerase ,Programmed cell death ,Skin Neoplasms ,Metastatic melanoma ,Mice, SCID ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Young Adult ,03 medical and health sciences ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Telomerase reverse transcriptase ,Melanoma ,neoplasms ,lcsh:QH301-705.5 ,Aged ,Telomere Homeostasis ,Middle Aged ,Telomere ,medicine.disease ,Molecular biology ,030104 developmental biology ,The Hallmarks of Cancer ,lcsh:Biology (General) ,Cell culture ,Lymphatic Metastasis ,Cancer research ,Female ,Neoplasm Transplantation - Abstract
Summary: Unlimited replicative potential is one of the hallmarks of cancer cells. In melanoma, hTERT (telomerase reverse transcriptase) is frequently overexpressed because of activating mutations in its promoter, suggesting that telomerase is necessary for melanoma development. We observed, however, that a subset of melanoma metastases and derived cell lines had no telomere maintenance mechanism. Early passages of the latter displayed long telomeres that progressively shortened and fused before cell death. We propose that, during melanoma formation, oncogenic mutations occur in precursor melanocytes with long telomeres, providing cells with sufficient replicative potential, thereby bypassing the need to re-activate telomerase. Our data further support the emerging idea that long telomeres promote melanoma formation. These observations are important when considering anticancer therapies targeting telomerase. : Viceconte et al. report that metastatic cutaneous melanoma cells display long telomeres and do not always have an active telomere maintenance mechanism, showing that the ability to maintain telomeres is not an absolute requirement for metastasis formation. These observations are important in the context of anti-cancer therapies targeting telomerase. Keywords: telomeres, telomerase, melanoma, ever-shorter telomeres, immortalization
- Published
- 2017
41. PL3.3 Second interim and first molecular analysis of the EORTC randomized phase III intergroup CATNON trial on concurrent and adjuvant temozolomide in anaplastic glioma without 1p/19q codeletion
- Author
-
Jean-François Baurain, Paul Clement, Michael Weller, Warren P. Mason, Helen Wheeler, Sara Erridge, Pim J. French, Kenneth Aldape, Wolfgang Wick, Mircea Tesileanu, Martin J. van den Bent, Brigitta G. Baumert, Alba A. Brandes, Anna K. Nowak, Marc Sanson, Michael A. Vogelbaum, Vassilis Golfinopoulos, Thierry Gorlia, Johan M. Kros, and Pieter Wesseling
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,O-6-methylguanine-DNA methyltransferase ,1p/19q Codeletion ,Interim analysis ,medicine.disease ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Interim ,Glioma ,Internal medicine ,medicine ,Oral Presentations ,Neurology (clinical) ,business ,Adjuvant ,030217 neurology & neurosurgery ,medicine.drug - Abstract
BACKGROUND The 1st interim analysis of the CATNON trial showed benefit from adjuvant (adj) temozolomide (TMZ) on overall survival (OS) but remained inconclusive about concurrent (conc) TMZ. A 2nd interim analysis was planned after 356 events. MATERIAL AND METHODS The 2x2 factorial design phase III CATNON trial randomized 751 adult patients with newly diagnosed non-codeleted anaplastic glioma to either 59.4 Gy radiotherapy (RT) alone; the same RT with concTMZ; the same RT and 12 cycles of adjTMZ or the same RT with both concTMZ and adjTMZ (doi: 10.1016/S0140-6736(17)31442-3). MGMT promoter methylation (MGMTmeth) status was re-assessed with the Infinium Methylation EPIC Beadchip using the MGMT_STP27 model. Isocitrate dehydrogenase 1 and 2 (IDH) mutation (mt) status was assessed with glioma targeted Agilent SureSelect baits sequence using an Illumina HiSeq2500 Rapid PE100. RESULTS With a median follow-up of 56 months and 356 events, the hazard ratio (HR) for OS adjusted for stratification factors after concTMZ was 0.968 (99.1% CI 0.73, 1.28). 5-year OS was 50.2% with and 52.7% without concTMZ (95% CI [44.4, 55.7] and [46.9, 58.1]). An IDHmt was found in 335 of 480 assessed cases (70%). Median OS was 19 mo (95% CI 16.3, 22.3) in IDHwt tumors and 116 mo (95% CI 82.0, 116.6) in IDHmt tumors. The interaction test based on IDH status was significant (p=0.016) in the univariate HR analysis for OS after concTMZ (IDHwt, n=145, events=120, HR = 1.27, 95% CI 0.89, 1.82, p=0.19; IDHmt, n=335, events=92, HR= 0.67, 95% CI 0.44, 1.03, p=0.06). IDHmt was predictive of benefit from adjTMZ (IDHmt HR: 0.41, 95% CI 0.27, 0.64; IDHwt: HR 1.05, 95% CI 0.73, 1.52; interaction test p = 0.001). In IDHmt patients that received adjTMZ, the HR for OS after concTMZ was 0.71 (95% CI 0.35, 1.42, p=0.32). MGMTmeth was found in 288 of 410 assessed cases (70%), interaction test for concTMZ (p = 0.092) and adjTMZ (p = 0.166) did not reach statistical significance. CONCLUSION In the entire study cohort, concTMZ did not increase OS. However, in IDHmt tumors a trend towards benefit of concTMZ is present. AdjTMZ increased OS in IDHmt but not in IDHwt tumors. Further analyses and follow-up will allow full assessment of efficacy in the molecular subgroups.
- Published
- 2019
42. A phase I study (E011-MEL) of a TriMix-based mRNA immunotherapy (ECI-006) in resected melanoma patients: Analysis of safety and immunogenicity
- Author
-
Annemie Rutten, Christof Vulsteke, Brenda De Keersmaecker, Javier Carrasco, Bart Neyns, Tim Van Assche, Ana Maria Arance Fernandez, Jean-François Baurain, Bertil Lindmark, Ainara Soria, Clinical sciences, Medical Oncology, Laboratory of Molecular and Medical Oncology, Laboratory of Molecullar and Cellular Therapy, and Basic (bio-) Medical Sciences
- Subjects
Cancer Research ,Messenger RNA ,CD40 ,biology ,business.industry ,medicine.medical_treatment ,Melanoma ,Immunogenicity ,Immunotherapy ,Dendritic cell ,medicine.disease ,Trimix ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,business ,030215 immunology ,CD70 - Abstract
2641 Background: ECI-006 is a combination of TriMix (mRNAs encoding for dendritic cell [DC] activating molecules [CD40L, CD70 and caTLR4]), and mRNAs encoding for melanoma-specific tumor-associated antigens (TAAs): tyrosinase, gp100, MAGE-A3, MAGE-C2, and PRAME. DCs transfected ex vivo with TriMix and TAAs mRNAs showed significant clinical activity in combination with ipilimumab in metastatic melanoma without increasing toxicity. This study aims to assess the safety and immunogenicity of ECI-006 vaccine administered intranodally (i.n.) in an adjuvant setting for patients with resected melanoma. Methods: Twenty patients who underwent resection of stage IIc/III/IV cutaneous melanoma received 5 administrations of ECI-006 (either 600 µg or 1800 µg [n = 10, each]) injected i.n. on Day 1 and after 2, 4, 6 and 14 weeks. Treatment-emergent adverse events (TEAEs) were graded using CTCAE version 4.0.3. Blood samples for immune monitoring (ELISPOT and intracellular cytokine staining [ICS]) were collected pre-dose and at weeks 4, 7, 14 and 15. Results: Nineteen patients completed the treatment. One patient in the low dose group discontinued the study after 4 doses due to disease relapse. Administration of ECI-006 was well tolerated. No serious adverse events or TEAEs Grade 3 or higher were reported. Of all TEAEs, myalgia and fatigue were the most reported in 3 (15%) and 5 (25%) patients, respectively. ELISPOT and ICS were performed on T cells pre-stimulated in vitro for 10-12 days, using a previously in-house validated protocol. Vaccine-induced immune responses according to predefined criteria were detected in 4/10 and 3/9 patients treated with the low and high dose, respectively. Samples from these patients are currently being subjected to T-cell receptor repertoire analysis. Conclusions: Among patients undergoing resection of stage IIc/III/IV melanoma, i.n. administration of ECI-006 at 600 or 1800 µg was generally well tolerated. ECI-006 demonstrated to be immunogenic in a proportion of patients. These results warrant further development of ECI-006 in combination with anti-PD-1 therapy in melanoma patients. Clinical trial information: NCT03394937.
- Published
- 2019
43. Phase II randomised discontinuation trial of brivanib in patients with advanced solid tumours
- Author
-
Peter J. O'Dwyer, Mark J. Ratain, Robert G. Maki, Stan B. Kaye, Charles M. Rudin, Gary K. Schwartz, David Khayat, Ahmad Awada, Joanna Vitfell-Rasmussen, Jennifer Obel, Maja DeJonge, Rebecca Kristeleit, Philippe L. Bedard, David R. D'Adamo, Ian B. Walters, Mark A. Dickson, Judith de Vos-Geelen, Samir D. Undevia, Jacek Jassem, Albiruni Ryan Abdul Razak, David Geary, Jacques Medioni, Jacques DeGreve, T.R. Jeffry Evans, Robin L. Jones, Michael B. Sawyer, Bruce Brockstein, Lillian L. Siu, Linda Janisch, Jean-François Baurain, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, UCL - (SLuc) Unité d'oncologie médicale, UCL - (SLuc) Centre du cancer, Medical Genetics, Clinical sciences, Laboratory of Molecular and Medical Oncology, Medical Oncology, Interne Geneeskunde, MUMC+: MA Medische Oncologie (9), and RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy
- Subjects
0301 basic medicine ,Male ,Cancer Research ,GROWTH-FACTOR RECEPTOR-2 ,Gastroenterology ,Withholding Treatment/statistics & numerical data ,0302 clinical medicine ,Neoplasms ,Randomised discontinuation trial ,Alanine ,Triazines ,Sarcomas ,INHIBITOR ,Alanine/analogs & derivatives ,Middle Aged ,Prognosis ,Survival Rate ,SOFT-TISSUE SARCOMA ,Transitional cell carcinoma ,Brivanib ,Oncology ,030220 oncology & carcinogenesis ,Female ,FGF2 status ,medicine.medical_specialty ,Antineoplastic Agents ,Placebo ,PAZOPANIB ,03 medical and health sciences ,Breast cancer ,Ovarian cancer ,Pancreatic cancer ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Lung cancer ,Aged ,Solid tumours ,business.industry ,Antineoplastic Agents/therapeutic use ,Cancer ,medicine.disease ,Triazines/therapeutic use ,Discontinuation ,Neoplasms/drug therapy ,Cancérologie ,030104 developmental biology ,Withholding Treatment ,Biomarkers, Tumor/metabolism ,business ,Follow-Up Studies - Abstract
Background: Brivanib is a selective inhibitor of vascular endothelial growth factor and fibroblast growth factor (FGF) signalling. We performed a phase II randomised discontinuation trial of brivanib in 7 tumour types (soft-tissue sarcomas [STS], ovarian cancer, breast cancer, pancreatic cancer, non-small-cell lung cancer [NSCLC], gastric/esophageal cancer and transitional cell carcinoma [TCC]). Patients and methods: During a 12-week open-label lead-in period, patients received brivanib 800 mg daily and were evaluated for FGF2 status by immunohistochemistry. Patients with stable disease at week 12 were randomised to brivanib or placebo. A study steering committee evaluated week 12 response to determine if enrolment in a tumour type would continue. The primary objective was progression-free survival (PFS) for brivanib versus placebo in patients with FGF2-positive tumours. Results: A total of 595 patients were treated, and stable disease was observed at the week 12 randomisation point in all tumour types. Closure decisions were made for breast cancer, pancreatic cancer, NSCLC, gastric cancer and TCC. Criteria for expansion were met for STS and ovarian cancer. In 53 randomised patients with STS and FGF2-positive tumours, the median PFS was 2.8 months for brivanib and 1.4 months for placebo (hazard ratio [HR]: 0.58, p = 0.08). For all randomised patients with sarcomas, the median PFS was 2.8 months (95% confidence interval [CI]: 1.4–4.0) for those treated with brivanib compared with 1.4 months (95% CI: 1.3–1.6) for placebo (HR = 0.64, 95% CI: 0.38–1.07; p = 0.09). In the 36 randomised patients with ovarian cancer and FGF2-positive tumours, the median PFS was 4.0 (95% CI: 2.6–4.2) months for brivanib and 2.0 months (95% CI: 1.2–2.7) for placebo (HR: 0.56, 95% CI: 0.26–1.22). For all randomised patients with ovarian cancer, the median PFS in those randomised to brivanib was 4.0 months (95% CI: 2.6–4.2) and was 2.0 months (95% CI: 1.2–2.7) in those randomised to placebo (HR = 0.54, 95% CI: 0.25–1.17; p = 0.11). Conclusion: Brivanib demonstrated activity in STS and ovarian cancer with an acceptable safety profile. FGF2 expression, as defined in the protocol, is not a predictive biomarker of the efficacy of brivanib., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2019
44. Co-primary endpoint of overall survival for tebentafusp (tebe)-induced rash in a phase 3 randomized trial comparing tebe versus investigator’s choice (IC) in first-line metastatic uveal melanoma
- Author
-
Anthony M. Joshua, Jessica C. Hassel, John M. Kirkwood, Sophie Piperno-Neumann, Christopher J. Holland, Richard D. Carvajal, Sebastian Ochsenreither, Alexander N. Shoushtari, Omid Hamid, Max Schlaak, Shaad Essa Abdullah, Jean-François Baurain, Reinhard Dummer, Ryan J. Sullivan, Marcus O. Butler, Howard Goodall, Piotr Rutkowski, Paul C. Nathan, Marlana Orloff, and Joseph J. Sacco
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Effector ,Melanoma ,First line ,T-cell receptor ,medicine.disease ,Rash ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Clinical endpoint ,Overall survival ,medicine.symptom ,business - Abstract
9527 Background: Tebe is a bispecific consisting of an affinity-enhanced T cell receptor fused to an anti-CD3 effector that can redirect T cells to target gp100+ cells. In this Phase (Ph) 3, randomized trial of first line (1L) metastatic uveal melanoma (mUM) [NCT03070392], tebe significantly improved overall survival (OS) vs. investigator’s choice (IC) in the intention-to-treat population (ITT). In previous trials, tebe-related skin adverse events (AEs), hypothesized to be on-target, off-tumor activity against gp100-expressing melanocytes, were associated with improved OS. This association was tested prospectively as a co-primary endpoint in the Ph3 study. Methods: 378 1L HLA-A*02:01+ mUM pts were randomized 2:1 to tebe (n = 252) or IC (n = 126). Co-primary endpoints were 1) OS in all randomized pts (ITT) and 2) OS in tebe-randomized pts who develop any grade rash in week (wk) 1 vs. all receiving IC. Rash was defined as composite of preferred AE terms. Melanocyte-related AEs (MRAEs) were defined as pigment change AEs in the skin or hair. Overall study-wide alpha was controlled at 0.05, with 90% assigned to ITT and 10% to rash. This analysis was conducted on the first interim analysis (data extracted Nov-2020). Results: In the 245 tebe treated pts, the characteristic skin related AEs included most frequently rash (at any time) in 201 pts (82%), pruritis in 167 pts (68%), MRAEs in 109 pts (45%) and erythema in 69 pts (28%). While rash, erythema and pruritis mostly occurred in the first 4 weeks, MRAEs occurred after a median of 2.7 mo. Rash captures most pts, 201/227 (89%), who have any of these skin related AEs. Rash occurred in 146 pts (60%) by wk 1; 179 pts (73%) by wk 2; and 195 pts (80%) by wk 3. Tebe pts with wk 1 rash had significantly longer OS vs. the IC arm, HR 0.35 (95% CI 0.23, 0.53), p < 0.0001. The estimated 1-yr OS rates were 83% vs 58%, respectively. When expanded to include tebe pts with rash through wk 3, the 1-yr OS rate of 75% was still numerically higher than IC. The 50 (20%) tebe pts who did not experience rash by week 3 had 1-yr OS rate of 55%. Conclusions: In 1L mUM pts, tebe significantly improved OS compared to IC in the ITT analysis. Week 1 rash, presumed due to tebe redirection of T cells to gp100+ skin melanocytes, was associated with a very strong OS benefit. Therefore, rash may be a marker that the immune system can be mobilized by tebe to target gp100+ cells. The vast majority of tebe pts will develop a rash at some point, and tebe pts without rash may still derive benefit. Clinical trial information: NCT03070392.
- Published
- 2021
45. Overall survival benefit from tebentafusp in patients with best response of progressive disease
- Author
-
John M. Kirkwood, Ryan J. Sullivan, Jessica C. Hassel, Shaad Essa Abdullah, Piotr Rutkowski, Sophie Piperno-Neumann, Joseph J. Sacco, Max Schlaak, Marcus O. Butler, Josep M. Piulats, Sebastian Ochsenreither, Jean-François Baurain, Anthony M. Joshua, Paul C. Nathan, Reinhard Dummer, Sarah Lockwood, Alexander N. Shoushtari, Marlana Orloff, and Mughda Deo
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,T-cell receptor ,macromolecular substances ,medicine.disease ,Internal medicine ,medicine ,Overall survival ,In patient ,business ,Progressive disease - Abstract
9509 Background: Tebentafusp (tebe) is the first T cell receptor (TCR) therapeutic to demonstrate an overall survival (OS) benefit in a randomized Phase 3 (Ph3) study [ NCT03070392 ]. In Ph2, 42% of pts with best overall response (BOR) of progressive disease (PD) survived > 1 year (yr), suggesting RECIST-based radiographic assessments underestimate OS benefit of tebe. Here we analyzed OS in the Ph3 study in a cohort of pts with BOR of PD by comparing tebe to the control arm of investigator’s choice (IC). Methods: 378 pts were randomized in a 2:1 ratio to tebe vs. IC. BOR was assessed by investigators using RECIST v1.1. Treatment beyond first disease progression (TBP) was permitted for both arms. On the IC arm, only patients receiving pembrolizumab (pembro) continued with TBP and were included in the TBP-related analyses. No crossover to tebe was permitted; investigators were free to choose subsequent therapy. This analysis was conducted on the first interim analysis (data extracted Nov-2020). Kaplan-Meier estimates of OS were based on Day 100 landmark to eliminate immortal time bias and to capture majority of the PDs. Results: By Day 100, PD as BOR occurred in 52% (130/252) of tebe pts (PD-tebe) vs. 60% (76/126) of IC pts (PD-IC). Key baseline characteristics including lactate dehydrogenase, alkaline phosphatase, ECOG performance, age, and sex were similar between PD-tebe vs PD-IC. The proportion of pts with PD due to progression of target lesions (TL), non-TL, or new lesions were also similar between the two groups. More pts received TBP among PD-tebe 53% (69/130) vs PD-pembro 16% (10/61). Median duration of TBP was longer for PD-tebe (7 weeks) vs PD-Pembro (3 weeks). The safety profile of PD-tebe pts during TBP was similar to all tebe-treated pts. OS was superior for PD-tebe vs PD-IC, HR = 0.41 (95%CI 0.25-0.66), even when considering key baseline covariates. While some pts had regression of TL despite diagnosis of PD ( < 10% of pts), the OS benefit remained even when limited to pts with best change of tumor growth of TL, HR 0.46 (0.29, 0.73). 58% (75/130) PD-tebe and 52% (40/76) PD-IC pts received subsequent therapies. In a landmark OS analysis of these pts beginning on 1st day of subsequent therapy, prior tebe was associated with better OS vs. prior IC, HR 0.59 (95%CI 0.36-0.96). Conclusions: Tebe is the first TCR therapeutic to demonstrate an OS benefit in a solid tumor. Surprisingly, a strong OS benefit from tebe is observed even in pts with BOR of PD, suggesting that RECIST-based radiographic assessments do not capture the complete benefit from tebe. The safety profile of tebe during TBP was consistent with that for long-term tebe treatment. Clinical trial information: NCT03070392.
- Published
- 2021
46. Interim Analysis of Phase 2 Results for Cemiplimab in Patients with Metastatic Basal Cell Carcinoma (mBCC) who Progressed on or are Intolerant to Hedgehog Inhibitors (HHIs)
- Author
-
Jean-François Baurain, Frank Seebach, Alexander J. Stratigos, Leonel Hernandez-Aya, Lisa Licitra, Aleksandar Sekulic, Katty Peris, Zeynep Eroglu, Marcus O. Butler, Ebony Coates, Israel Lowy, Oliver Bechter, Timothy Bowler, Anne Lynn S. Chang, Kosalai Kal Mohan, Karl D. Lewis, Emmanuel Okoye, Emily S. Ruiz, Matthew G. Fury, Lara Dunn, Siyu Li, Rogerio I. Neves, Suk-Young Yoo, Axel Hauschild, and Michael R. Migden
- Subjects
business.industry ,Cancer research ,Medicine ,In patient ,Interim analysis ,business ,Hedgehog ,Metastatic basal cell carcinoma - Abstract
not available.
- Published
- 2021
47. Evaluation of an individualized starting-dose of niraparib in the PRIMA/ENGOT-OV26/GOG-3012 study
- Author
-
Lydia Gaba, Mansoor Raza Mirza, Sharad A. Ghamande, Oi Wah S. Yap, Divya Gupta, David M. O'Malley, Bradley J. Monk, Emily Prendergast, Hannelore Denys, Philippe Follana, Jean-François Baurain, Antonio Gonzalez Martin, Peter G. Rose, Whitney Graybill, Jacob Korach, Paula Calvert, Carmela Pisano, Klaus Baumann, Eva Guerra, and Y. Li
- Subjects
Oncology ,03 medical and health sciences ,Cancer Research ,medicine.medical_specialty ,0302 clinical medicine ,Recurrent Ovarian Cancer ,business.industry ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,030215 immunology - Abstract
6050 Background: Niraparib is approved at a fixed starting dose (FSD) of 300 mg QD for maintenance treatment of patients (pts) with recurrent ovarian cancer (OC) achieving a complete or partial response to platinum-based chemotherapy based in the ENGOT-OV16/NOVA study. A post-hoc analysis of NOVA showed baseline bodyweight (BW) and platelet count (PC) were predictive for hematologic toxicities and dose reductions. Following this analysis, the PRIMA/ENGOT-OV26/GOG-3012 study was amended to prospectively evaluate the safety and efficacy of an individualized starting dose (ISD) regimen. Methods: This double-blind, placebo-controlled, phase III study randomized 733 pts with newly diagnosed advanced OC with a complete or partial response to first-line (1L) platinum-based chemotherapy. The protocol was amended to change the dose from 300 mg FSD for all patients to an ISD regimen: 200 mg QD in pts with BW
- Published
- 2020
48. Randomized phase II study of sapanisertib (SAP) + paclitaxel (PAC) versus PAC alone in patients (pts) with advanced, recurrent, or persistent endometrial cancer
- Author
-
Robert M. Wenham, Douglas V. Faller, Francesco Raspagliesi, Vicky Makker, Amit M. Oza, Nicoletta Colombo, Giovanni Scambia, Jean-François Baurain, Zhenqiang Su, Katherine M. Moxley, Rachel Neuwirth, Ana Oaknin, Jonathan Krell, Andrea Jewell, Sileny Han, Farhad Sedarati, Elena Ioana Braicu, Sylvie Vincent, and Eva Guerra
- Subjects
Cancer Research ,Endometrial Tumor ,endocrine system diseases ,business.industry ,Endometrial cancer ,education ,Dual inhibitor ,food and beverages ,Phases of clinical research ,medicine.disease ,humanities ,chemistry.chemical_compound ,Oncology ,Paclitaxel ,chemistry ,health services administration ,Cancer research ,Medicine ,In patient ,business ,Sapanisertib - Abstract
6087 Background: SAP (TAK-228, MLN0128) is a selective dual inhibitor of mammalian target of rapamycin complexes 1 and 2. In endometrial tumor xenograft models, SAP+PAC exhibited stronger antitumor efficacy than PAC alone. Methods: Female pts with histologic/cytologic diagnosis of endometrial cancer were randomized to receive SAP 4 mg by mouth (days [d] 2–4, 9–11, 16–18, 23–25) plus PAC 80 mg/m2 intravenously (d 1, 8, 15), or PAC alone, in 28-day cycles until unacceptable toxicity or disease progression. Randomization was stratified by histologic subtype, lines of prior chemotherapy (1 vs. 2), and prior taxane therapy. The primary endpoint was progression-free survival (PFS); secondary endpoints included overall survival (OS), overall response rate (ORR), clinical benefit rate (CBR; ORR + stable disease), and safety. Additional treatment arms of SAP alone (weekly dosing) and SAP+TAK-117 were closed after futility analyses. Results: 180 pts were randomized to SAP+PAC (n=90) or PAC (n=90); 86 and 87 pts received SAP+PAC and PAC, respectively; 3 pts from each arm were ongoing on treatment at data cut (30 July 2019). Baseline characteristics were balanced between arms. After a median follow-up of 17.2 vs. 14.4 mos with SAP+PAC vs. PAC, median PFS was 5.6 mos vs. 3.7 mos (hazard ratio [HR] 0.82; 95% CI 0.58–1.15). In pts with endometrioid histology (n=116), median PFS was 5.7 mos with SAP+PAC vs 3.3 mos with PAC (HR 0.66; 95% CI 0.43–1.03). In pts with nonendometrioid histology (n=64), median PFS was 3.6 mos with SAP+PAC vs. 5.4 mos with PAC (HR 1.09; 95% CI 0.62–1.90). Median OS was 13.7 mos with SAP+PAC vs. 14.6 mos with PAC (HR 1.01; 95% CI 0.67–1.53). Confirmed ORR was 24% with SAP+PAC vs. 18% with PAC (endometrioid, 23% vs. 16%; nonendometrioid, 28% vs. 22%); CBR was 80% vs. 58% (endometrioid, 84% vs. 55%; nonendometrioid, 72% vs. 63%). Median number of cycles received was 5 (range 1–23) with SAP+PAC and 4 (range 1–37) with PAC. Rates of grade ≥3 treatment-emergent adverse events (TEAEs) were 90% with SAP+PAC vs. 54% with PAC; the most common included anemia (21% vs.12%), neutropenia (12% vs. 3%), fatigue (12% vs. 5%), hypophosphatemia (12% vs. 1%), and pulmonary embolism (11% vs. 3%). Conclusions: Median PFS was longer with SAP+PAC vs. PAC in pts with endometrial cancer but did not reach statistical significance. PFS was particularly longer in the endometrioid subtype but again was not significant, and further studies are warranted. Incidence of grade ≥3 TEAEs was higher with SAP+PAC vs. PAC, but SAP+PAC toxicity was manageable, with no new safety signals. Clinical trial information: NCT02725268.
- Published
- 2020
49. Association of homogeneous inflamed gene signature with a better outcome in patients with metastatic melanoma treated with MAGE-A3 immunotherapeutic
- Author
-
Jamila Louahed, Silvija Jarnjak, Frederic Lehmann, Florent Grange, Bart Neyns, Fernando Ulloa-Montoya, Marc Gillet, Caroline Robert, Jean-François Baurain, Pedro Miguel De Sousa Alves, Céleste Lebbé, Laurent Mortier, Clinical sciences, Medical Oncology, Laboratory of Molecular and Medical Oncology, and Laboratory of Molecullar and Cellular Therapy
- Subjects
0301 basic medicine ,safety ,Cancer Research ,medicine.medical_specialty ,endocrine system ,mage-a3 immunotherapeutic ,Phases of clinical research ,Gastroenterology ,gene signature ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Internal medicine ,medicine ,melanoma ,Adverse effect ,Original Research ,biology ,business.industry ,Immunogenicity ,Melanoma ,Gene signature ,medicine.disease ,Vaccination ,clinical activity ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,business - Abstract
Purpose This study assessed clinical activity, safety and immunogenicity of MAGE-A3 immunotherapeutic in patients with MAGE-A3-positive metastatic melanoma. Patients and methods In this open-label, multicentre, uncontrolled, Phase II study (ClinicalTrials.gov NCT00896480),patients received ≤24 doses of MAGE-A3 immunotherapeutic (4-cycle schedule). At screening, two skin lesions were biopsied for MAGE-A3 expression analysis and presence/absence of a previously identified gene signature (GS) associated with favourable clinical outcome. Clinical activity was assessed in terms of clinical response, time-to-treatment failure (TTF) and progression-free survival (PFS). Adverse events (AEs) and serious AEs (SAEs) were recorded. MAGE-A3-specific immune responses were assessed. Clinical activity and immunogenicity were analysed overall and separately in patients with 2/2 (GS+/+), 1/2 (GS+/-) or 0/2 (GS-/-) biopsies presenting GS. Results Of 49 screened patients, 32 had MAGE-A3-positive tumours; 24 (8 GS+/+, 8 GS+/-, 8 GS-/-) were treated. Two complete (GS+/+ patients) and two partial responses (one GS+/+, one GS+/-) were reported; of note, one of the two complete responses was unlikely to be related to the study treatment. Median TTF and PFS were 14.8 and 7.2 months for GS+/+, 2.3 and 2.8 months for GS+/- and 2.4 and 2.9 months for GS-/- patients. Three grade 3 AEs and two SAEs unrelated to treatment were reported. All patients were seropositive for MAGE-A3 antibodies on vaccination with no differences between the different GS profiles. MAGE-A3-specific CD4+ and CD8+ T cell immunogenicity was detected; 12/16 (75.0%) of patients presented CD4+ T cell responses. Conclusion Treatment with MAGE-A3 immunotherapeutic showed signs of clinical activity in GS+/+ patients. Treatment was well tolerated and immunogenic. No differences in immune responses according to GS status were observed. Trial registration number NCT00896480 (Results).
- Published
- 2018
50. Fertility sparing treatgment in patients with early stage endometrial cancer, using a combination of surgery and GnRH agonists: a monocentric retrospective study and review of the literature
- Author
-
Pascale Jadoul, Mathieu Luyckx, Etienne Marbaix, Jean-Luc Squifflet, Stéphanie Tock, Jean-François Baurain, UCL - SSS/IREC/GYNE - Pôle de Gynécologie, UCL - (SLuc) Service de gynécologie et d'andrologie, UCL - (SLuc) Unité d'oncologie médicale, UCL - (SLuc) Service d'anatomie pathologique, UCL - SSS/DDUV - Institut de Duve, UCL - SSS/DDUV/CELL - Biologie cellulaire, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, and UCL - (SLuc) Centre du cancer
- Subjects
fertility-sparing ,medicine.medical_specialty ,Atypical endometrial hyperplasia ,medicine.medical_treatment ,endometrial adenocarcinoma ,conservative treatment ,Endometrial adenocarcinoma ,GnRH agonist ,atypical endometrial hyperplasia ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Carcinoma ,Radical surgery ,Atypical Endometrial Hyperplasia ,Original Research ,lcsh:R5-920 ,030219 obstetrics & reproductive medicine ,Hysterectomy ,medicine.diagnostic_test ,business.industry ,Fertility-sparing ,Endometrial cancer ,Retrospective cohort study ,General Medicine ,medicine.disease ,Surgery ,Pregnancy rate ,Hysteroscopy ,030220 oncology & carcinogenesis ,Medicine ,lcsh:Medicine (General) ,business ,Conservative treatment - Abstract
Objectives: To evaluate the efficacy and safety of gonadotropin-releasing hormone (GnRH) agonist after endometrial resection in women suffering early stage endometrial carcinoma (EC) and/or endometrial intra-epithelial neoplasia (EIN). Design: A retrospective review of clinical files between January 1999 and December 2016. Setting: University hospital. Patients: Eighteen women younger than 41 years with grade 1 endometrial carcinoma (G1EC) and/or Endometrial intra-epithelial neoplasia (EIN). Interventions: All patients received GnRH agonist for 3 months after an endometrial resection combined with a laparoscopy to exclude concomitant ovarian tumor and/or other extra-uterine disease. The patient underwent a follow-up of 3 months interval with endometrial sampling by hysteroscopy. Main Outcome Measure(s): The recurrence rate and the pregnancy rate after fertility sparing treatment. Results: We identified 9 patients with EIN (50%), 7 patients with G1EC (38.9%), 1 with combined histology (5.5%), and 1 with G2EC (5.5%). After a median follow-up of 40.7 months, 12 patients conserved their uterus (66.7%), and 8 (53.3%) patients were pregnant with a total of 14 pregnancies among those who tried to become pregnant. We observed a complete response rate in 12 patients (66.7%) but 3 of these patients relapsed (25%). We also found a stable disease in 6 patients (33.3%). Conclusions: Compared with other fertility sparing treatments, GnRH agonist after surgery is an effective fertility-sparing strategy for women with EIN and/or G1EC. We recommend hysterectomy once a family has been completed even if the literature does not clearly lead to radical surgery.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.