105 results on '"Vasiliki Pelekanou"'
Search Results
2. 1276 Leveraging artificial intelligence (AI) models delineating tumor vs immune cell expression for scalable biomarker analysis of clinical trial samples: a digital image analysis approach for NSCLC
- Author
-
David Schaer, Rosemarie Krupar, Vasiliki Pelekanou, Yuko Ishii, Simon Schallenberg, Sharon Ruane, Cornelius Böhm, Lukas Ruff, Maximilian Alber, Katja Lingelbach, Blanca Pablos, Simon Heinke, Nader Aldoj, Suhas Pandhe, Luca Pizzagalli, Maximilian Gottschalk, Bartosz Wójtowicz, Stephan Tietz, Marion Rudolph, and Emmanuelle di Tomaso
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. AMEERA-1 phase 1/2 study of amcenestrant, SAR439859, in postmenopausal women with ER-positive/HER2-negative advanced breast cancer
- Author
-
Aditya Bardia, Sarat Chandarlapaty, Hannah M. Linden, Gary A. Ulaner, Alice Gosselin, Sylvaine Cartot-Cotton, Patrick Cohen, Séverine Doroumian, Gautier Paux, Marina Celanovic, Vasiliki Pelekanou, Jeffrey E. Ming, Nils Ternès, Monsif Bouaboula, Joon Sang Lee, Anne-Laure Bauchet, and Mario Campone
- Subjects
Science - Abstract
There is a need for potent and non-toxic estrogen receptor (ER) antagonists to overcome the limitations of existing endocrine therapies. Here the authors report the results from Arm 1 of the Phase 1/2 study (AMEERA-1) among postmenopausal women with ER+/HER2− advanced breast cancer, which evaluates the safety, antitumor activity, pharmacokinetics, and pharmacodynamics of amcenestrant administered as monotherapy.
- Published
- 2022
- Full Text
- View/download PDF
4. Tumor immune microenvironment of self-identified African American and non-African American triple negative breast cancer
- Author
-
Michal Marczyk, Tao Qing, Tess O’Meara, Vesal Yagahoobi, Vasiliki Pelekanou, Yalai Bai, Emily Reisenbichler, Kimberly S. Cole, Xiaotong Li, Vignesh Gunasekharan, Eiman Ibrahim, Kristina Fanucci, Wei Wei, David L. Rimm, Lajos Pusztai, and Kim R. M. Blenman
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Differences in the tumor immune microenvironment may result in differences in prognosis and response to treatment in cancer patients. We hypothesized that differences in the tumor immune microenvironment may exist between African American (AA) and NonAA patients, due to ancestry-related or socioeconomic factors, that may partially explain differences in clinical outcomes. We analyzed clinically matched triple-negative breast cancer (TNBC) tissues from self-identified AA and NonAA patients and found that stromal TILs, PD-L1 IHC-positivity, mRNA expression of immune-related pathways, and immunotherapy response predictive signatures were significantly higher in AA samples (p
- Published
- 2022
- Full Text
- View/download PDF
5. Quantitative assessment of the immune microenvironment in African American Triple Negative Breast Cancer: a case–control study
- Author
-
Vesal Yaghoobi, Myrto Moutafi, Thazin Nwe Aung, Vasiliki Pelekanou, Sanam Yaghoubi, Kim Blenman, Eiman Ibrahim, Ioannis A. Vathiotis, Saba Shafi, Anup Sharma, Tess O’Meara, Aileen I. Fernandez, Lajos Pusztai, and David L. Rimm
- Subjects
Triple-negative breast cancer ,Tumor microenvironment ,Quantitative immunofluorescence ,African American ,Immune cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Purpose Triple negative breast cancer (TNBC) is more common in African American (AA) than Non-AA (NAA) population. We hypothesize that tumor microenvironment (TME) contributes to this disparity. Here, we use multiplex quantitative immunofluorescence to characterize the expression of immunologic biomarkers in the TME in both populations. Patients and methods TNBC tumor resection specimen tissues from a 100-patient case: control cohort including 49 AA and 51 NAA were collected. TME markers including CD45, CD14, CD68, CD206, CD4, CD8, CD20, CD3, Ki67, GzB, Thy1, FAP, aSMA, CD34, Col4, VWF and PD-L1 we quantitatively assessed in every field of view. Mean expression levels were compared between cases and controls. Results Although no significant differences were detected in individual lymphoid and myeloid markers, we found that infiltration with CD45+ immune cells (p = 0.0102) was higher in TNBC in AA population. AA TNBC tumors also had significantly higher level of lymphocytic infiltration defined as CD45+ CD14− cells (p = 0.0081). CD3+ T-cells in AA tumors expressed significantly higher levels of Ki67 (0.0066) compared to NAAs, indicating that a higher percentage of AA tumors contained activated T-cells. All other biomarkers showed no significant differences between the AA and NAA group. Conclusions While the TME in TNBC is rich in immune cells in both racial groups, there is a numerical increase in lymphoid infiltration in AA compared to NAA TNBC. Significantly, higher activated T cells seen in AA patients raises the possibility that there may be a subset of AA patients with improved response to immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
6. AMEERA-5: a randomized, double-blind phase 3 study of amcenestrant plus palbociclib letrozole plus palbociclib for previously untreated ER+/HER2– advanced breast cancer
- Author
-
Aditya Bardia, Javier Cortes, Sara A. Hurvitz, Suzette Delaloge, Hiroji Iwata, Zhi-Ming Shao, Dheepak Kanagavel, Patrick Cohen, Qianying Liu, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, and Joyce O’Shaughnessy
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: For estrogen receptor–positive (ER+)/human epidermal growth factor receptor 2–negative (HER2–) advanced breast cancer (ABC), the current standard first-line treatment includes an aromatase inhibitor in combination with a cyclin-dependent kinase 4/6 inhibitor. When resistance occurs, often related to the occurrence of ESR1 mutations, selective estrogen receptor modulators or degraders (SERDs) may be used, alone or in combination regimens. Amcenestrant (SAR439859), an optimized oral SERD, has shown clinical antitumor activity in combination with palbociclib in patients with ER+/HER2– ABC and, as monotherapy, in patients with and without ESR1 mutations. Here, we describe the study design of AMEERA-5, an ongoing, prospective, phase 3, randomized, double-blind, multinational study comparing the efficacy and safety of amcenestrant plus palbociclib versus letrozole plus palbociclib in patients with advanced (locoregional recurrent or metastatic) ER+/HER2– breast cancer. Methods: Patients are pre-/postmenopausal women and men with no prior systemic therapy for ABC. The planned enrollment is 1066 patients. Patients are randomized 1:1 to either amcenestrant 200 mg plus palbociclib 125 mg or letrozole 2.5 mg plus palbociclib 125 mg. Amcenestrant, letrozole, and their matching placebos are taken once daily continuously; palbociclib is taken once daily for 21 days, followed by 7 days off-treatment for a 28-day cycle. Treatment continues until disease progression, unacceptable toxicity, or decision to stop treatment. Pre-/perimenopausal women and men receive goserelin subcutaneously. Randomization is stratified by de novo metastatic disease, menopausal status, and visceral metastases. The primary endpoint is progression-free survival. The key secondary endpoint is overall survival; others are safety, pharmacokinetics, and quality of life. Conclusions: AMEERA-5 is evaluating the efficacy and safety of amcenestrant in combination with palbociclib as first-line therapy in pre-/postmenopausal women and men with ER+/HER2– ABC. ClinicalTrials Identifier: NCT04478266.
- Published
- 2022
- Full Text
- View/download PDF
7. Immune profiling of pre- and post-treatment breast cancer tissues from the SWOG S0800 neoadjuvant trial
- Author
-
Xiaotong Li, Sarah Warren, Vasiliki Pelekanou, Vikram Wali, Alessandra Cesano, Mingdong Liu, Patrick Danaher, Nathane Elliott, Zeina A. Nahleh, Daniel F. Hayes, Gabriel N. Hortobagyi, William E. Barlow, Christos Hatzis, and Lajos Pusztai
- Subjects
Tumor infiltrating lymphocytes ,PD-L1 ,Immune-related genes ,Neoadjuvant treatment ,Bevacizumab ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background How the immune microenvironment changes during neoadjuvant chemotherapy of primary breast cancer is not well understood. Methods We analyzed pre- and post-treatment samples from 60 patients using the NanoString PanCancer IO360™ assay to measure the expression of 750 immune-related genes corresponding to 14 immune cell types and various immune functions, and assessed TIL counts and PD-L1 protein expression by immunohistochemistry. Treatment associated changes in gene expression levels were compared using t-test with Bonferroni correction. TIL count, PD-L1 protein and immune metagenes were compared using Wilcoxon test. Baseline immune markers were correlated with pathologic complete response (pCR) using estrogen receptor and treatment arm adjusted logistic regression. Results At baseline, high TIL counts and high expression of chemoattractant cytokines (CCL21, CCL19) and cytotoxic T cell markers were associated with higher pCR rate. High expression of stromal genes (VEGFB, TGFB3, PDGFB, FGFR1, IGFR1), mast and myeloid inflammatory cell metagenes, stem cell related genes (CD90, WNT11, CTNNB1) and CX3CR1, and IL11RA were associated with residual disease (RD). After treatment, in cases with pCR, TIL counts and most immune genes decreased significantly. Among RD cases, TIL counts and PD-L1 expression did not change but cellular stress and hypoxia associated genes (DUSP1, EGR1), and IL6, CD36, CXCL2, CD69 and the IL8/VEGF metagene increased. Conclusions Activated T cells in the tumor microenvironment are associated with pCR whereas stromal functions are associated with residual disease. Most immune functions decrease during neoadjuvant chemotherapy but several immunotherapy targets (PD-L1, IL6, IL8) remain expressed in RD suggesting potential therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
8. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers
- Author
-
Vasiliki Pelekanou, Franz Villarroel-Espindola, Kurt A. Schalper, Lajos Pusztai, and David L. Rimm
- Subjects
Tumor-associated macrophages ,Matrix metalloproteinase-9 ,Breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The role of tumor-associated macrophages (TAMs) in the cancer immune landscape and their potential as treatment targets or modulators of response to treatment are gaining increasing interest. TAMs display high molecular and functional complexity. Therefore their objective assessment as breast cancer biomarkers is critical. The aims of this study were to objectively determine the in situ expression and significance of TAM biomarkers (CD68, CD163, and MMP-9) in breast cancer and to identify subclasses of patients who could benefit from TAM-targeting therapies. Methods We measured CD68, CD163, and MMP-9 protein expression in formalin-fixed paraffin-embedded tissues of breast carcinomas represented in tissue microarray format using multiplexed quantitative immunofluorescence (QIF) in two independent Yale cohorts: cohort A—n = 398, estrogen receptor–positive (ER+) and ER− cases—and the triple-negative breast cancer (TNBC)-only cohort B (n = 160). Associations between macrophage markers, ER status, and survival were assessed. Protein expression measured by QIF was compared with mRNA expression data from the METABRIC study. Results All three macrophage markers were co-expressed, displaying higher expression in ER− cancers. High pan-macrophage marker CD68 correlated with poorer overall survival (OS) only in ER− cases of cohort A (P = 0.02). High expression of CD163 protein in TAMs was associated with improved OS in ER− cases (cohort A, P = 0.03 and TNBC cohort B, P = 0.04, respectively) but not in ER+ cancers. MMP-9 protein was not individually associated with OS. High expression of MMP-9 in the CD68+/CD163+ TAMs was associated with worse OS in ER+ tumors (P
- Published
- 2018
- Full Text
- View/download PDF
9. Androgen Triggers the Pro-Migratory CXCL12/CXCR4 Axis in AR-Positive Breast Cancer Cell Lines: Underlying Mechanism and Possible Implications for the Use of Aromatase Inhibitors in Breast Cancer
- Author
-
Kalliopi Azariadis, Fotini Kiagiadaki, Vasiliki Pelekanou, Vasiliki Bempi, Kostas Alexakis, Marilena Kampa, Andreas Tsapis, Elias Castanas, and George Notas
- Subjects
Androgen ,Androgen Receptor ,CXCL12 ,NCOA1 ,CXCR4 ,P53 ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background/Aims: Reports regarding the role of androgen in breast cancer (BC) are conflicting. Some studies suggest that androgen could lead to undesirable responses in the presence of certain BC tumor characteristics. We have shown that androgen induces C-X-C motif chemokine 12 (CXCL12) in BC cell lines. Our aim was to identify the mechanisms regulating the phenotypic effects of androgen-induced CXCL12 on Androgen Receptor (AR) positive BC cell lines. Methods: We analyzed the expression of CXCL12 and its receptors with qPCR and ELISA and the role of Nuclear Receptor Coactivator 1 (NCOA1) in this effect. AR effects on the CXCL12 promoter was studied via Chromatin-immunoprecipitation. We also analyzed publically available data from The Cancer Genome Atlas to verify AR-CXCL12 interactions and to identify the effect or Aromatase Inhibitors (AI) therapy on CXCL12 expression and disease progression in AR positive cases. Results: CXCL12 induction occurs only in AR-positive BC cell lines, possibly via an Androgen Response Element, upstream of the CXCL12 promoter. The steroid receptor co-regulator NCOA1 is critical for this effect. Androgen only induced the motility of p53-mutant BC cells T47D cells via upregulation of CXCR4 expression while they had no effect on wild-type p53 MCF-7 cells. Loss of CXCR4 expression and depletion of CXCL12 abolished the effect of androgen in T47D cells while inhibition of p53 expression in MCF-7 cells made them responsive to androgen and increased their motility in the presence to androgen. Patients with estrogen receptor positive (ER+)/AR+ BC treated with AIs were at increased risk of disease progression compared to ER+/AR+ non-AI treated and ER+/AR- AI treated cases. Conclusion: AIs may lead to unfavorable responses in some ER/AR positive BC cases, especially in patients with AR+, p53 mutant tumors.
- Published
- 2017
- Full Text
- View/download PDF
10. Effect of neoadjuvant chemotherapy on tumor-infiltrating lymphocytes and PD-L1 expression in breast cancer and its clinical significance
- Author
-
Vasiliki Pelekanou, Daniel E. Carvajal-Hausdorf, Mehmet Altan, Brad Wasserman, Cristobal Carvajal-Hausdorf, Hallie Wimberly, Jason Brown, Donald Lannin, Lajos Pusztai, and David L. Rimm
- Subjects
Tumor infiltrating lymphocytes ,Programmed death ligand 1 ,Neoadjuvant treatment ,Breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The effects of neoadjuvant chemotherapy on immune markers remain largely unknown. The specific aim of this study was to assess stromal tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) protein expression in a cohort of breast cancer patients treated with neoadjuvant chemotherapy. Methods Using quantitative immunofluorescence, we investigated stromal TILs and PD-L1 protein expression in pre-treatment and residual breast cancer tissue from a Yale Cancer Center patient cohort of 58 patients diagnosed with breast cancer from 2003 to 2009 and treated with neoadjuvant chemotherapy. We compared the TIL count and PD-L1 status in paired pre-treatment and residual cancer tissues and correlated changes and baseline levels with survival. Results Of the 58 patients, 46 (79.3%) had hormone-positive and 34 (58.6%) had node-positive breast cancer. Eighty-six percent of residual cancer tissues had TIL infiltration and 17% had PD-L1 expression. There was a trend for higher TIL counts in postchemotherapy compared to prechemotherapy samples (p = 0.09). Increase in TIL count was associated with longer 5-year recurrence-free survival (p = 0.02, HR = 3.9, 95% CI = 1.179–15.39). PD-L1 expression (both stromal and tumor cells) was significantly lower in post-treatment samples (p = 0.001). Change in PD-L1 expression after therapy or TILs and PD-L1 expression in the posttreatment samples did not correlate with survival. Conclusions Increase in stromal TILs in residual cancer compared to pretreatment tissue is associated with improved recurrence-free survival. Despite a trend for increasing TIL counts, PD-L1 expression decreased in residual disease compared to pretreatment samples.
- Published
- 2017
- Full Text
- View/download PDF
11. BCMA (TNFRSF17) Induces APRIL and BAFF Mediated Breast Cancer Cell Stemness
- Author
-
Vasiliki Pelekanou, George Notas, Paraskevi Athanasouli, Konstantinos Alexakis, Fotini Kiagiadaki, Nikolaos Peroulis, Konstantina Kalyvianaki, Errika Kampouri, Hara Polioudaki, Panayiotis Theodoropoulos, Andreas Tsapis, Elias Castanas, and Marilena Kampa
- Subjects
APRIL (TNFSF13) ,BAFF (TNFSF13B) ,BCMA (TNFRSF17) ,pluripotency ,cancer stem cells ,therapy resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Recent advances in cancer immunology revealed immune-related properties of cancer cells as novel promising therapeutic targets. The two TNF superfamily members, APRIL (TNFSF13), and BAFF (TNFSF13B), which are type II membrane proteins, released in active forms by proteolytic cleavage and are primarily involved in B-lymphocyte maturation, have also been associated with tumor growth and aggressiveness in several solid tumors, including breast cancer. In the present work we studied the effect of APRIL and BAFF on epithelial to mesenchymal transition, migration, and stemness of breast cancer cells. Our findings show that both molecules increase epithelial to mesenchymal transition and migratory capacity of breast cancer cells, as well as cancer stem cell numbers, by increasing the expression of pluripotency genes such as ALDH1A1, KLF4, and NANOG. These effects are mediated by their common receptor BCMA (TNFRSF17) and the JNK signaling pathway. Interestingly, transcriptional data analysis from breast cancer cells and patients revealed that androgens can increase APRIL transcription and subsequently, in an autocrine/paracrine manner, enhance its pluripotency effect. In conclusion, our data suggest a possible role of APRIL and BAFF in breast cancer disease progression and provide evidence for a new possible mechanism of therapy resistance, that could be particularly relevant in aromatase inhibitors-treated patients, were local androgen is increased.
- Published
- 2018
- Full Text
- View/download PDF
12. Erratum to: Effect of neoadjuvant chemotherapy on tumor-infiltrating lymphocytes and PD-L1 expression in breast cancer and its clinical significance
- Author
-
Vasiliki Pelekanou, Daniel E. Carvajal-Hausdorf, Mehmet Altan, Brad Wasserman, Cristobal Carvajal-Hausdorf, Hallie Wimberly, Jason Brown, Donald Lannin, Lajos Pusztai, and David L. Rimm
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2017
- Full Text
- View/download PDF
13. Abstract P1-17-11: Updated data from AMEERA-1: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), combined with palbociclib in postmenopausal women with ER+/HER2- advanced breast cancer
- Author
-
Sarat Chandarlapaty, Hannah M Linden, Patrick Neven, Katarina Petrakova, Aditya Bardia, Peter Kabos, Sofia Braga, Valentina Boni, Alice Gosselin, Marina Celanovic, Patrick Cohen, Gautier Paux, Vasiliki Pelekanou, Nils Ternès, Joon Sang Lee, and Mario Campone
- Subjects
Cancer Research ,Oncology - Abstract
Background: In Arm 2 of the ongoing AMEERA-1 trial (NCT03284957), amcenestrant, an optimized oral SERD combined with the CDK4/6 inhibitor (CDK4/6i) palbociclib demonstrated favorable safety and encouraging antitumor activity among patients with endocrine-resistant ER+/HER2− advanced breast cancer in dose escalation (Part C) and dose expansion (Part D) (Chandarlapaty et al., ASCO 2021; abstract 1058). Here we report an update of safety, antitumor activity data, and progression-free survival (PFS), of amcenestrant 200 mg in combination with palbociclib. Analysis of genomic data, including modulation over time and correlation with clinical outcome, will also be presented. Methods: The trial enrolled postmenopausal women with ER+/HER2- locally-advanced or metastatic breast cancer with disease progression while on ≥ 6 months of prior endocrine therapy (ET) in the advanced setting, or who relapsed on adjuvant ET after the first 2 years of treatment or within 12 months of completing adjuvant ET. Prior chemotherapy (≤ 1) was allowed as well as prior CDK4/6i-based therapy (≤ 1, in Part C only). In this pooled analysis (N = 39), patients in Parts C + D received amcenestrant 200 mg once daily + palbociclib 125 mg (21 days on/7 days off), administered in 28-day cycles. Safety in the pooled analysis was reported using methods previously described (Chandarlapaty et al., ASCO 2021; abstract 1058). Data from investigator-assessed, response-evaluable patients in the pooled analysis without prior exposure to targeted therapies (N = 34) were used to evaluate antitumor activity per RECIST v1.1, including the objective response rate (ORR), clinical benefit rate (CBR), and PFS. Results: At a data cutoff of May 30, 2021, in the pooled analysis (N = 39), the median (range) duration of treatment exposure was 44.3 weeks (1-80). Of 39 patients, 24 (61.5%) had initiated at least 10 cycles (40 weeks) of treatment, with 20/39 (51.3%) still receiving ongoing treatment. Among the 34/39 (87.2%) patients in the response-evaluable population, median follow-up was 48.3 weeks with a PFS probability of being event free at 24 weeks of 78.2% (95% CI: 59.6%; 89.0%). Median PFS is not yet mature, with 14/34 (41.2%) patients having had a PFS event (all were progression events and no deaths occurred). The ORR was 11/34 (32.4%; all partial responses). Clinical benefit at 24 weeks was seen in 25/34 (CBR = 73.5%) patients. Median (range) time to first response was 16.3 weeks (8-32). Amcenestrant treatment-related adverse events (TRAEs) and palbociclib TRAEs, respectively, occurred in 27/39 (69.2%) and 35/39 (89.7%) patients for all grade events and in 5/39 (12.8%) and 18/39 (46.2%) patients for Grade ≥ 3 events. Non-hematological amcenestrant and palbociclib TRAEs are reported in Table 1. Neutrophil count decrease based on hematological laboratory abnormalities was observed in the majority of patients (94.9%; with Grade ≥ 3 in 56.4%). Conclusions: Among postmenopausal women with endocrine-resistant ER+/HER2- advanced breast cancer, amcenestrant 200 mg in combination with the approved dose of palbociclib continues to demonstrate encouraging long-term antitumor activity, sustained clinical benefit, and a favorable safety profile consistent with previous results. Funding: Sanofi. Table 1.Non-hematological amcenestrant and palbociclib TRAEs occurring in > 10% of patientsPooled Analysis. Amcenestrant 200 mg + Palbociclib. (Parts C + D; N = 39)Amcenestrant Non-hematological TRAEs, n (%)All GradesGrade ≥ 3–Fatigue7 (17.9)0–Nausea7 (17.9)0–Arthralgia4 (10.3)0–Asthenia4 (10.3)0–Hot flush4 (10.3)0Palbociclib Non-hematological TRAEs, n (%)All GradesGrade ≥ 3–Fatigue12 (30.8)0–Nausea10 (25.6)0–Asthenia4 (10.3)0–Dysgeusia4 (10.3)0–Stomatitis4 (10.3)0 Citation Format: Sarat Chandarlapaty, Hannah M Linden, Patrick Neven, Katarina Petrakova, Aditya Bardia, Peter Kabos, Sofia Braga, Valentina Boni, Alice Gosselin, Marina Celanovic, Patrick Cohen, Gautier Paux, Vasiliki Pelekanou, Nils Ternès, Joon Sang Lee, Mario Campone. Updated data from AMEERA-1: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), combined with palbociclib in postmenopausal women with ER+/HER2- advanced breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P1-17-11.
- Published
- 2022
- Full Text
- View/download PDF
14. Abstract OT2-11-04: Ameera-1 Arm 5: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with abemaciclib in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer
- Author
-
Mario Campone, Sarat Chandarlapaty, Aditya Bardia, Patrick Neven, Katarina Petrakova, Peter Kabos, Valentina Boni, Sofia Braga, Marina Celanovic, Patrick Cohen, Alice Gosselin, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, and Hannah Linden
- Subjects
Cancer Research ,Oncology - Abstract
Background Endocrine therapy in combination with a targeted cyclin-dependent kinase (CDK) 4/6 inhibitor is the clinical standard for treatment of ER+/HER2- advanced breast cancer. Amcenestrant (SAR439859) is an optimized oral SERD with potent dual activity that antagonizes and degrades the ER, resulting in inhibition of the ER signaling pathway. In previous arms of the AMEERA-1 study, amcenestrant, as monotherapy or in combination with the CDK4/6 inhibitor palbociclib, demonstrated antitumor activity and a favorable safety profile in postmenopausal women with heavily pretreated ER+/HER2- advanced breast cancer. The objective of Arm 5 of the AMEERA-1 study is to evaluate safety and antitumor activity of amcenestrant in combination with the CDK4/6 inhibitor abemaciclib for patients with ER+/HER2- advanced breast cancer. Methods AMEERA-1 (NCT03284957) is an open-label, non-comparative, dose escalation and dose expansion Phase 1/2 study of amcenestrant as monotherapy, then in combination with other anti-cancer targeted therapies. Arm 5 investigates dose escalation (Part J) and dose expansion (Part K), of amcenestrant in combination with abemaciclib. Postmenopausal women with ER+/HER2- advanced breast cancer, ECOG performance status 0-1, and ≥ 6 months prior endocrine therapy are eligible. In Arm 5 (Parts J and K), ≤ 1 prior line of a single endocrine therapy for advanced disease is allowed. Prior treatment with fulvestrant or any other SERD is not allowed; in addition, prior therapy with CDK4/6 inhibitors for advanced disease is not allowed. Part J allows ≤ 1 prior chemotherapy for advanced disease, while prior chemotherapy for advanced disease is not allowed in Part K. Additional exclusion criteria in Arm 5 are prior drugs targeting the phosphoinositide 3-kinase axis; history of or concurrent pneumonitis; and history of or concurrent venous thromboembolism (i.e., deep vein thrombosis, pulmonary embolism, or cerebral venous sinus thrombosis). Part J evaluates the selected amcenestrant dose for combination therapy plus abemaciclib 150 mg twice daily (BID) (the approved standard dose) or abemaciclib 100 mg BID, taken in 28-day cycles. Additional dose levels of amcenestrant may be explored based on safety and pharmacokinetics (PK). The objective of Part J is to determine the recommended dose (RD) of abemaciclib in combination with the selected amcenestrant dose for combination therapy, based on preliminary safety, PK, and antitumor activity data. The primary endpoint in Part J is the incidence of treatment-related dose-limiting toxicities (DLTs) at Cycle 1. Approximately up to 12 DLT-evaluable patients will be needed to establish the RD of abemaciclib in combination with amcenestrant in Part J. In Part K, approximately 20 patients will be treated at the RD of abemaciclib for combination therapy with amcenestrant, the primary endpoint being safety and tolerability. Secondary endpoints include PK and antitumor activity. Funding: Sanofi. Citation Format: Mario Campone, Sarat Chandarlapaty, Aditya Bardia, Patrick Neven, Katarina Petrakova, Peter Kabos, Valentina Boni, Sofia Braga, Marina Celanovic, Patrick Cohen, Alice Gosselin, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, Hannah Linden. Ameera-1 Arm 5: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with abemaciclib in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr OT2-11-04.
- Published
- 2022
- Full Text
- View/download PDF
15. Abstract OT2-11-03: AMEERA-1 : Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with alpelisib in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-)PIK3CA-mutated advanced breast cancer
- Author
-
Mario Campone, Aditya Bardia, Peter Kabos, Sarat Chandarlapaty, Patrick Neven, Valentina Boni, Simon Lord, Sylvaine Cartot-Cotton, Marina Celanovic, Alice Gosselin, Vasiliki Pelekanou, and Hannah M Linden
- Subjects
Cancer Research ,Oncology - Abstract
Background Amcenestrant is an optimized oral SERD with potent dual activity of ER antagonism and degradation resulting in inhibition of ER signaling. Amcenestrant monotherapy or combination with palbociclib showed antitumor activity and a favorable safety profile in postmenopausal women with heavily pretreated ER+/HER2- mBC. PIK3CA mutations are associated with endocrine resistance in ER+/HER2- patients (pts). Published data support the addition of the PI3Kα inhibitor alpelisib to SERD therapy for these pts. Methods AMEERA-1 (NCT03284957) is an open-label, non-comparative, dose escalation and dose expansion Phase 1/2 study of amcenestrant as monotherapy, then in combination with other anti-cancer targeted therapies. Parts F and G investigate safety run-in and dose expansion, respectively, of amcenestrant in combination with alpelisib. Postmenopausal women with ER+/HER2- advanced breast cancer, PIK3CA mutated in tumor tissue or cfDNA, ECOG performance status 0-1, and ≥ 6 months prior endocrine therapy are eligible. Pts must have progressed on an aromatase inhibitor plus CDK4/6 inhibitor as first-line therapy for advanced disease. Part F allows ≤ 1 prior chemotherapy for advanced disease; no prior chemotherapy is allowed in Part G. Exclusion criteria in Parts F and G include prior drugs targeting the PI3K axis, type 1 diabetes, uncontrolled type 2 diabetes, history of severe cutaneous reactions, and ongoing osteonecrosis of the jaw. Part F assesses dose-limiting toxicities and pharmacokinetics (PK) of a standard dose of amcenestrant plus the approved dose of alpelisib (300 mg once daily). Additional amcenestrant doses or a lower dose of alpelisib may be explored based on safety and PK. The primary objective in Part F is to confirm the recommended phase 2 dose (RP2D) of amcenestrant in combination with alpelisib, based on safety. In Part G, approximately 34 pts will be treated at the RP2D, the primary endpoint being safety and tolerability. Secondary endpoints include PK and antitumor activity. This study is currently recruiting participants. This abstract was previously submitted to the 2021 European Society for Medical Oncology Annual Congress. Funding: Sanofi. Citation Format: Mario Campone, Aditya Bardia, Peter Kabos, Sarat Chandarlapaty, Patrick Neven, Valentina Boni, Simon Lord, Sylvaine Cartot-Cotton, Marina Celanovic, Alice Gosselin, Vasiliki Pelekanou, Hannah M Linden. AMEERA-1 : Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with alpelisib in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-)PIK3CA-mutated advanced breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr OT2-11-03.
- Published
- 2022
- Full Text
- View/download PDF
16. Abstract OT2-11-02: Ameera-1 Arm 4: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with everolimus in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer
- Author
-
Mario Campone, Patrick Neven, Katarina Petrakova, Sofia Braga, Marina Celanovic, Patrick Cohen, Alice Gosselin, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, and Valentina Boni
- Subjects
Cancer Research ,Oncology - Abstract
Background Amcenestrant (SAR439859) is an optimized oral SERD with potent dual activity that antagonizes and degrades the ER, resulting in inhibition of the ER signaling pathway. Amcenestrant, as monotherapy or in combination with palbociclib, has shown antitumor activity and a favorable safety profile in postmenopausal women with heavily pretreated ER+/HER2- advanced breast cancer. Published data support the addition of targeted therapy to endocrine therapy for patients with ER+/HER2- advanced breast cancer. The objective of Arm 4 of the AMEERA-1 study is to evaluate safety and antitumor activity of amcenestrant in combination with the mammalian target of rapamycin (mTOR) inhibitor everolimus for patients with ER+/HER2- advanced breast cancer. Methods AMEERA-1 (NCT03284957) is an open-label, non-comparative, dose escalation and dose expansion Phase 1/2 study of amcenestrant as monotherapy, then in combination with other anti-cancer targeted therapies. Arm 4 investigates dose escalation (Part H) and dose expansion (Part I), of amcenestrant in combination with everolimus. Postmenopausal women with ER+/HER2- advanced breast cancer, ECOG performance status 0-1, and ≥ 6 months prior endocrine therapy are eligible. In Arm 4 (Parts H and I), ≤ 1 prior line of a single endocrine therapy for advanced disease is allowed. Patients must have progressed on a non-steroidal aromatase inhibitor plus cyclin-dependent kinase 4/6 inhibitor as first-line therapy for advanced disease. Prior treatment with fulvestrant or any other SERD is not allowed. Part H allows ≤ 1 prior chemotherapy for advanced disease; no prior chemotherapy for advanced disease is allowed in Part I. Exclusion criteria in Arm 4 include prior drugs targeting the phosphoinositide 3-kinase axis; history of or concurrent pneumonitis; history of severe cutaneous reactions; type 1 diabetes; uncontrolled type 2 diabetes; uncontrolled hypercholesterolemia, hypertriglyceridemia, and hyperglycemia; any uncontrolled infection; uncontrolled stomatitis, angioedema due to angiotensin-converting enzyme inhibitors, and impaired wounds. Part H evaluates the selected amcenestrant dose for combination therapy plus everolimus 10 mg once daily (QD) (the approved standard dose) or everolimus 5 mg QD, taken in 28-day cycles. Additional amcenestrant doses may be explored based on safety and pharmacokinetics (PK). The objective of Part H is to determine the recommended dose (RD) of everolimus in combination with the selected amcenestrant dose for combination therapy, based on preliminary safety, PK, and antitumor activity data. The primary endpoint in Part H is the incidence of treatment-related dose-limiting toxicities (DLTs) at Cycle 1. Approximately up to 12 DLT-evaluable patients will be needed to establish the RD of everolimus in combination with amcenestrant in Part H. In Part I, approximately 12 patients will be treated at the RD of everolimus for combination therapy with amcenestrant, the primary endpoint being safety and tolerability. Secondary endpoints include PK and antitumor activity. Funding: Sanofi. Citation Format: Mario Campone, Patrick Neven, Katarina Petrakova, Sofia Braga, Marina Celanovic, Patrick Cohen, Alice Gosselin, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, Valentina Boni. Ameera-1 Arm 4: Phase 1/2 study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with everolimus in postmenopausal women with ER+/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr OT2-11-02.
- Published
- 2022
- Full Text
- View/download PDF
17. Abstract OT2-11-08: AMEERA-5 : A randomized, double-blind phase 3 study of amcenestrant (SAR439859) + palbociclib versus letrozole + palbociclib for previously untreated ER+/HER2- advanced breast cancer
- Author
-
Aditya Bardia, Javier Cortes, Sara Hurvitz, Suzette Delaloge, Hiroji Iwata, Zhi-Ming Shao, Dheepak Kanagavel, Patrick Cohen, Qianying Liu, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, and Joyce O’Shaughnessy
- Subjects
Cancer Research ,Oncology - Abstract
Background Selective estrogen receptor degraders (SERDs) block estrogen receptor (ER) associated signaling and have created interest for treating patients (pts) with advanced ER+ breast cancer (BC). Fulvestrant is currently the only SERD available for advanced BC but requires intramuscular administration, limiting the applied dose, exposure and receptor engagement. Amcenestrant (SAR439859) is an oral SERD that binds with high affinity to both wild-type and mutant ER, blocking estradiol binding and promoting up to 98% ER degradation in preclinical studies. In the phase I AMEERA-1 study of pretreated pts with ER+/HER2- advanced BC, amcenestrant 150-600 mg once daily (QD) showed a mean ER occupancy of 94% with plasma concentrations > 100 ng/mL and a favorable safety profile (Bardia, 2019; data on file). Combination therapy with amcenestrant + palbociclib (palbo) was also evaluated as part of this ongoing phase I study. CDK 4/6 inhibitors (CDK4/6i) combined with an aromatase inhibitor (AI), the gold standard for first line treatment for advanced breast cancer, prolong progression free survival (PFS) in pts with no prior treatment for ER+/HER2- advanced BC, but OS benefit has not been shown yet in postmenopausal pts. There remains a clinical need for more effective treatments in this setting. Methods AMEERA-5 (NCT04478266) is an ongoing, prospective, randomized, double-blind phase III study comparing the efficacy and safety of amcenestrant + palbo with that of letrozole + palbo in pts with advanced, locoregional recurrent or metastatic ER+/HER2- BC who have not received prior systemic therapy for advanced disease. The study includes men, pre/peri-menopausal (with goserelin) and post-menopausal women. Pts with progression during or within 12 months of (neo)adjuvant endocrine therapy using any of the following agents are excluded: AI, selective estrogen receptor modulators, CDK4/6i. Pts are randomized 1:1 to either continuous amcenestrant 200 mg or letrozole 2.5 mg QD orally with matching placebos; both combined with palbo 125 mg QD orally (d1-21 every 28-d cycle). Randomization is stratified according to disease type (de novo metastatic vs recurrent disease), the presence of visceral metastasis, and menopausal status. The primary endpoint is investigator assessed progression free survival (PFS) (RECIST v1.1). Secondary endpoints are overall survival, PFS2, objective response rate, duration of response, clinical benefit rate, pharmacokinetics of amcenestrant and palbo, health-related quality of life, time to chemotherapy, and safety. Biomarkers will be measured in paired tumor biopsies and cell free deoxyribonucleic acid (cfDNA) over time. Target enrolment = 1066 pts; enrolment as of 6/2021 = 415 pts. Bardia A, et al., J Clin Oncol. 2019; 37 (15 suppl):1054. Funding: Sanofi. This abstract was accepted and previously presented at the 2021 American Society of Clinical Oncology Annual Meeting. All rights reserved. Citation Format: Aditya Bardia, Javier Cortes, Sara Hurvitz, Suzette Delaloge, Hiroji Iwata, Zhi-Ming Shao, Dheepak Kanagavel, Patrick Cohen, Qianying Liu, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, Joyce O’Shaughnessy. AMEERA-5 : A randomized, double-blind phase 3 study of amcenestrant (SAR439859) + palbociclib versus letrozole + palbociclib for previously untreated ER+/HER2- advanced breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr OT2-11-08.
- Published
- 2022
- Full Text
- View/download PDF
18. Data from B7-H3 Expression in NSCLC and Its Association with B7-H4, PD-L1 and Tumor-Infiltrating Lymphocytes
- Author
-
David L. Rimm, Roy S. Herbst, Konstantinos Syrigos, Patricia Gaule, Maria Toki, Kurt A. Schalper, Vasiliki Pelekanou, and Mehmet Altan
- Abstract
Purpose: The immune checkpoint PD-1 and its receptor B7-H1 (PD-L1) are successful therapeutic targets in cancer but less is known about other B7 family members. Here, we determined the expression level of B7-H3 protein in non–small cell lung cancer (NSCLC) and evaluated its association with tumor-infiltrating lymphocytes (TIL), PD-L1, B7-H4, and major clinicopathologic characteristics is in 3 NSCLC cohorts.Experimental design: We used multiplexed automated quantitative immunofluorescence (QIF) to assess the levels of B7-H3, PD-L1, B7-H4, and TILs in 634 NSCLC cases with validated antibodies. Associations between the marker levels, major clinicopathologic variables and survival were analyzed.Results: Expression of B7-H3 protein was found in 80.4% (510/634) of the cases. High B7-H3 protein level (top 10 percentile) was associated with poor overall survival (P < 0.05). Elevated B7-H3 was consistently associated with smoking history across the 3 cohorts, but not with sex, age, clinical stage, and histology. Coexpression of B7-H3 and PD-L1 was found in 17.6% of the cases (112/634) and with B7-H4 in 10% (63/634). B7-H4 and PD-L1 were simultaneously detected only in 1.8% of NSCLCs (12/634). The expression of B7-H3 was not associated with the levels of CD3-, CD8-, and CD20-positive TILs.Conclusions: B7-H3 protein is expressed in the majority of NSCLCs and is associated with smoking history. High levels of B7-H3 protein have a negative prognostic impact in lung carcinomas. Coexpression of B7-H3 with PD-L1 and B7-H4 is relatively low, suggesting a nonredundant biological role of these targets. Clin Cancer Res; 23(17); 5202–9. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figures 1-5, Supplementary Tables 1-2 from B7-H3 Expression in NSCLC and Its Association with B7-H4, PD-L1 and Tumor-Infiltrating Lymphocytes
- Author
-
David L. Rimm, Roy S. Herbst, Konstantinos Syrigos, Patricia Gaule, Maria Toki, Kurt A. Schalper, Vasiliki Pelekanou, and Mehmet Altan
- Abstract
Table 1. Clinico-pathologic features of the Non-small cell lung cancer cohorts (Cohort A, B and C); Table 2. Cox regression model with clinicopathologic features, tumor B7-H3, B7-H4 and PD-L1 protein expression risk ratios for 3 cohorts; Figure 1. B7-H3 antibody validation; Figure 2. B7-H4 antibody validation; Figure 3. B7-H3 tumor and stromal protein expression regression, staining examples; Figure 4. Co-expression co-localization of B7-H3 and PD-L1; Figure 5. Additional images for Co-expression co-localization of B7-H3 and B7-H4
- Published
- 2023
- Full Text
- View/download PDF
20. supp material from Biomarker Discovery in Patients with Immunotherapy-Treated Melanoma with Imaging Mass Cytometry
- Author
-
David L. Rimm, Harriet M. Kluger, Kurt A. Schalper, Brian Bourke-Martin, Vasiliki Pelekanou, Thazin Nwe Aung, Maria I. Toki, Pok Fai Wong, Franz Villarroel-Espindola, and Sandra Martinez-Morilla
- Abstract
supp material
- Published
- 2023
- Full Text
- View/download PDF
21. Biomarker Discovery in Patients with Immunotherapy-Treated Melanoma with Imaging Mass Cytometry
- Author
-
Thazin Nwe Aung, Franz Villarroel-Espindola, Pok Fai Wong, David L. Rimm, Sandra Martinez-Morilla, Vasiliki Pelekanou, Kurt A. Schalper, Brian Bourke-Martin, Maria I. Toki, and Harriet M. Kluger
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Microarray ,medicine.medical_treatment ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Humans ,Multiplex ,Mass cytometry ,RNA, Messenger ,Biomarker discovery ,Immune Checkpoint Inhibitors ,Melanoma ,Image Cytometry ,Tumor microenvironment ,business.industry ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,beta 2-Microglobulin ,business - Abstract
Purpose: Imaging mass cytometry (IMC) is among the first tools with the capacity for multiplex analysis of more than 40 targets, which provides a novel approach to biomarker discovery. Here, we used IMC to characterize the tumor microenvironment of patients with metastatic melanoma who received immunotherapy in efforts to find indicative factors of treatment response. In spite of the new power of IMC, the image analysis aspects are still limited by the challenges of cell segmentation. Experimental Design: Here, rather than segment, we performed image analysis using a newly designed version of the AQUA software to measure marker intensity in molecularly defined compartments: tumor cells, stroma, T cells, B cells, and macrophages. IMC data were compared with quantitative immunofluorescence (QIF) and digital spatial profiling. Results: Validation of IMC results for immune markers was confirmed by regression with additional multiplexing methods and outcome assessment. Multivariable analyses by each compartment revealed significant associations of 12 markers for progression-free survival and seven markers for overall survival (OS). The most compelling indicative biomarker, beta2-microglobulin (B2M), was confirmed by correlation with OS by QIF in the discovery cohort and validated in an independent published cohort profiled by mRNA expression. Conclusions: Using digital image analysis based on pixel colocalization to assess IMC data allowed us to quantitively measure 25 markers simultaneously on formalin-fixed, paraffin-embedded tissue microarray samples. In addition to showing high concordance with other multiplexing technologies, we identified a series of potentially indicative biomarkers for immunotherapy in metastatic melanoma, including B2M.
- Published
- 2021
- Full Text
- View/download PDF
22. Development of machine learning–powered models for prostate cancer HRD prediction
- Author
-
Michael Nercessian, Jake Conway, Vasiliki Pelekanou, Andreas Schlicker, Ekaterina Nevedomskaya, Darren Fahy, Julia Varao, Michael Pyle, Michael Drage, Archit Khosla, Benjamin Glass, Emmanuelle DiTomaso, and Yinghui Zhou
- Subjects
Cancer Research ,Oncology - Abstract
206 Background: In prostate cancer, homologous recombination deficiency (HRD) is associated with poor prognosis, and sensitivity to DNA damaging agents and DNA damage repair (DDR) inhibitors. As new classes of DDR inhibitors become available, identifying patients with HRD will be critical for treatment selection. Here, we present machine learning (ML)-based models trained to predict HRD status directly from hematoxylin and eosin (H&E) whole slide images (WSI). Methods: ML models were trained to predict and segment cells and tissue regions within the tumor microenvironment (TME) using annotated (N=91,021 annotations) WSI of H&E-stained resections from the cancer genome atlas prostate adenocarcinoma (TCGA PRAD) dataset (N=401) and needle core biopsies from a proprietary dataset (N=1,000). Quantified Human Interpretable Features (HIFs) that describe the TME composition were extracted. Three models were trained to predict HRD status using 373 WSI with known HRD score (TCGA PRAD; train N=259, validation N=76, and test N=38). Two models used input from the TME model: An HIF multivariate logistic regression model, and a graph neural network (GNN) where predictions are based on the complex spatial relationships within the TME. An end-to-end (E2E) multiple instance learning model predicted directly from the WSI. Two cutoffs for HRD were defined using Gaussian Mixture Models, resulting in 99 WSI (train N=72, validation N=18, and test N=9) positive for the Genomic Instability (>16 events) cutoff, and 58 WSI (train N=44, validation N=10, test N=4) positive for the Genomic Instability (>22 events) cutoff. An independent validation set of 45 biopsies and 16 resections from a biobank of metastatic castration resistant prostate cancer with HRD status determined by whole-exome sequencing was compared to ML model H&E-based HRD prediction. Results: In the TCGA test set of resection samples, all three models moderately or strongly predicted HRD status, with the HIF model showing the best performance (AUROC 0.87, sensitivity 0.88, specificity 0.62). The same HIF model performed equally well (AUROC 0.85, Sensitivity 0.93, specificity 0.67) in the resection samples from the independent validation set. However, the model performance went down (AUROC 0.69, sensitivity 0.91, specificity: 0.3) when both resection and needle biopsy samples were included, highlighting the importance of a representative training set to achieve robust performance in a real world setting. Further model training and validation with a more diverse dataset is required to accurately assess the performance of the model on needle biopsies. Conclusions: ML models trained on resection prostate cancer samples performed well in predicting HRD status when applied to the same sample type, demonstrating the potential of ML models to predict genomic biomarkers status in surgical specimens for treatment decision.
- Published
- 2023
- Full Text
- View/download PDF
23. Genomic and Immune Profiling of a Patient With Triple-Negative Breast Cancer That Progressed During Neoadjuvant Chemotherapy Plus PD-L1 Blockade
- Author
-
Xiaotong Li, Lajos Pusztai, Natalia Buza, Christos Hatzis, David Casadevall, Francesc López-Giráldez, Ryan L Powles, Vasiliki Pelekanou, Borbála Székely, Arjun Dhawan, Julia Foldi, and Vikram B. Wali
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,biology ,business.industry ,medicine.medical_treatment ,Case Report ,Blockade ,Immune profiling ,Text mining ,Internal medicine ,PD-L1 ,medicine ,biology.protein ,business ,Triple-negative breast cancer - Published
- 2019
- Full Text
- View/download PDF
24. Estrogen receptor-alpha isoforms are the main estrogen receptors expressed in non-small cell lung carcinoma
- Author
-
Marilena Kampa, Katerina Lavredaki, Efstathios N. Stathopoulos, Vasiliki Pelekanou, Efstathia Bakogeorgou, Elias Castanas, Eleni Moustou, Georgia Chinari, V. Georgoulias, Eleftheria Anastasiou, George Notas, Anthony O'Grady, Andreas Tsapis, P. Arapantoni, and Rolando Garcia-Milian
- Subjects
Adult ,Male ,0301 basic medicine ,Gene isoform ,Lung Neoplasms ,Clinical Biochemistry ,Cell ,Estrogen receptor ,Biology ,Biochemistry ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Protein Isoforms ,Receptor ,Molecular Biology ,Aged ,Retrospective Studies ,Aged, 80 and over ,Pharmacology ,Organic Chemistry ,Estrogen Receptor alpha ,Middle Aged ,Immunohistochemistry ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Estrogen receptor alpha ,Tamoxifen ,medicine.drug - Abstract
The expression profile of estrogen receptors (ER) in Non-Small Cell Lung Carcinoma (NSCLC) remains contradictory. Here we investigated protein and transcriptome expression of ERα wild type and variants. Tissue Micro-Arrays of 200 cases of NSCLC (paired tumor/non-tumor) were assayed by immunohistochemistry using a panel of ERα antibodies targeting different epitopes (HC20, 6F11, 1D5, ERα36 and ERα17p). ERβ epitopes were also examined for comparison. In parallel we conducted a probe-set mapping (Affymetrix HGU133 plus 2 chip) meta-analysis of 12 NSCLC tumor public transcriptomic studies (1418 cases) and 39 NSCLC cell lines. Finally, we have investigated early transcriptional effects of 17β-estradiol, 17β-estradiol-BSA, tamoxifen and their combination in two NSCLC cell lines (A549, H520). ERα transcript and protein detection in NSCLC specimens and cell lines suggests that extranuclear ERα variants, like ERα36, prevail, while wild-type ERα66 is minimally expressed. In non-tumor lung, the wild-type ERα66 is quasi-absent. The combined evaluation of ERα isoform staining intensity and subcellular localization with sex, can discriminate NSCLC subtypes and normal lung. Overall ERα transcription decreases in NSCLC. ERα expression is sex-related in non-tumor tissue, but in NSCLC it is exclusively correlating with tumor histologic subtype. ERα isoform protein expression is higher than ERβ. ERα isoforms are functional and display specific early transcriptional effects following steroid treatment. In conclusion, our data show a wide extranuclear ERα-variant expression in normal lung and NSCLC that is not reported by routine pathology ER evaluation criteria, limited in the nuclear wild type receptor.
- Published
- 2019
- Full Text
- View/download PDF
25. 333TiP AMEERA-1: Phase I/II study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with alpelisib in postmenopausal women with ER+/ human epidermal growth factor receptor 2-negative (HER2–) PIK3CA-mutated advanced breast cancer
- Author
-
Hannah M. Linden, Simon Lord, S. Cartot-Cotton, Vasiliki Pelekanou, A. Gosselin, M. Campone, Sarat Chandarlapaty, Patrick Neven, Aditya Bardia, Valentina Boni, Peter Kabos, and Marina Celanovic
- Subjects
Postmenopausal women ,Phase i ii ,Oncology ,business.industry ,Advanced breast ,medicine ,Cancer research ,Estrogen receptor ,Cancer ,Hematology ,medicine.disease ,business ,Human Epidermal Growth Factor Receptor 2 - Published
- 2021
- Full Text
- View/download PDF
26. 264P AMEERA-1: Subgroup analyses of phase I/II study of amcenestrant (SAR439859), an oral selective estrogen receptor (ER) degrader (SERD), with palbociclib in postmenopausal women with ER+/ human epidermal growth factor receptor 2-negative (HER2–) advanced breast cancer (aBC)
- Author
-
N. Ternes, Vasiliki Pelekanou, Sofia Braga, Patrick Neven, A. Gosselin, Hannah M. Linden, P. Cohen, M. Campone, G. Paux, Sarat Chandarlapaty, Aditya Bardia, Valentina Boni, Peter Kabos, K. Petrakova, and Marina Celanovic
- Subjects
Postmenopausal women ,business.industry ,Advanced breast ,Estrogen receptor ,Cancer ,Hematology ,Palbociclib ,medicine.disease ,Phase i ii ,Oncology ,Cancer research ,medicine ,business ,Human Epidermal Growth Factor Receptor 2 - Published
- 2021
- Full Text
- View/download PDF
27. Immunological Differences Between Immune-Rich Estrogen Receptor–Positive and Immune-Rich Triple-Negative Breast Cancers
- Author
-
Tess O'Meara, Tao Qing, Michal Marczyk, Lajos Pusztai, Kimberly Cole, Vesal Yaghoobi, Vasiliki Pelekanou, David L. Rimm, and Kim Blenman
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,business.industry ,Estrogen receptor ,chemical and pharmacologic phenomena ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Immune system ,Oncology ,Estrogen ,030220 oncology & carcinogenesis ,Original Reports ,medicine ,Cancer research ,business ,Triple negative - Abstract
PURPOSE A subset of estrogen receptor–positive (ER-positive) breast cancer (BC) contains high levels of tumor-infiltrating lymphocytes (TILs), similar to triple-negative BC (TNBC). The majority of immuno-oncology trials target TNBCs because of the greater proportion of TIL-rich TNBCs. The extent to which the immune microenvironments of immune-rich ER-positive BC and TNBC differ is unknown. PATIENTS AND METHODS RNA sequencing data from The Cancer Genome Atlas (TCGA; n = 697 ER-positive BCs; n = 191 TNBCs) were used for discovery; microarray expression data from Molecular Taxonomy of Breast Cancer International Consortium (METABRIC; n = 1,186 ER-positive BCs; n = 297 TNBCs) was used for validation. Patients in the top 25th percentile of a previously published total TIL metagene score distribution were considered immune rich. We compared expression of immune cell markers, immune function metagenes, and immuno-oncology therapeutic targets among immune-rich subtypes. RESULTS Relative fractions of resting mast cells (TCGA Padj = .009; METABRIC Padj = 4.09E-15), CD8+ T cells (TCGA Padj = .015; METABRIC Padj = 0.390), and M2-like macrophages (TCGA Padj= 4.68E-05; METABRIC Padj = .435) were higher in immune-rich ER-positive BCs, but M0-like macrophages (TCGA Padj = 0.015; METABRIC Padj = .004) and M1-like macrophages (TCGA Padj = 9.39E-08; METABRIC Padj = 6.24E-11) were higher in immune-rich TNBCs. Ninety-one immune-related genes (eg, CXCL14, CSF3R, TGF-B3, LRRC32/GARP, TGFB-R2) and a transforming growth factor β (TGF-β) response metagene were significantly overexpressed in immune-rich ER-positive BCs, whereas 41 immune-related genes (eg, IFNG, PD-L1, CTLA4, MAGEA4) were overexpressed in immune-rich TNBCs in both discovery and validation data sets. TGF-β pathway member genes correlated negatively with expression of immune activation markers ( IFNG, granzyme-B, perforin) and positively with M2-like macrophages ( IL4, IL10, and MMP9) and regulatory T-cell ( FOXP3) markers in both subtypes. CONCLUSION Different immunotherapy strategies may be optimal in immune-rich ER-positive BC and TNBC. Drugs targeting the TGF-β pathway and M2-like macrophages are promising strategies in immune-rich ER-positive BCs to augment antitumor immunity.
- Published
- 2020
28. Prospective multi-institutional evaluation of pathologist assessment of PD-L1 assays for patient selection in triple negative breast cancer
- Author
-
Kamaljeet Singh, Lajos Pusztai, Jane E. Brock, Anja C. Roden, Krisztina Z. Hanley, Gang Han, Hannah Wen, Dylan V. Miller, Mirna Podoll, Beth T. Harrison, Emily Reisenbichler, Shi Wei, Omar Hameed, Kimberly Cole, Fahad Shabbir Ahmed, Vasiliki Pelekanou, Andrew M. Bellizzi, M. Gabriela Kuba, David L. Rimm, Vesal Yaghoobi, Veerle Bossuyt, Amy Ly, Oluwole Fadare, and Mary Ann Sanders
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Intraclass correlation ,Concordance ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Article ,B7-H1 Antigen ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,PD-L1 ,Biomarkers, Tumor ,Medicine ,Humans ,In patient ,Prospective Studies ,Lung cancer ,Triple-negative breast cancer ,Reproducibility ,biology ,business.industry ,Patient Selection ,Reproducibility of Results ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Female ,business - Abstract
The US Food and Drug Administration (FDA) approved the PD-L1 immunohistochemical assay, SP142, as a companion test to determine eligibility for atezolizumab therapy in patients with advanced triple negative breast cancer (TNBC) but data in lung cancer studies suggest the assay suffers from poor reproducibility. We sought to evaluate reproducibility and concordance in PD-L1 scoring across multiple pathologists. Full TNBC sections were stained with SP142 and SP263 assays and interpreted for percentage (%) immune cell (IC) staining by 19 pathologists from 14 academic institutions. Proportion of PD-L1 positive cases (defined as ≥1% IC) was determined for each assay as well as concordance across observers. We utilized a new method we call Observers Needed to Evaluate Subjective Tests (ONEST) to determine the minimum number of evaluators needed to estimate concordance between large numbers of readers, as occurs in the real-world setting. PD-L1 was interpreted as positive with the SP142 assay in an average 58% of cases compared with 78% with SP263 (p
- Published
- 2020
29. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers
- Author
-
David L. Rimm, Kurt A. Schalper, Franz Villarroel-Espindola, Vasiliki Pelekanou, and Lajos Pusztai
- Subjects
0301 basic medicine ,medicine.drug_class ,Breast Neoplasms ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Surgical oncology ,Tumor Microenvironment ,Medicine ,Humans ,Breast ,Tissue microarray ,business.industry ,CD68 ,Tumor-associated macrophages ,Cancer ,Matrix metalloproteinase-9 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030104 developmental biology ,Matrix Metalloproteinase 9 ,Estrogen ,030220 oncology & carcinogenesis ,Cohort ,Cancer research ,business ,CD163 ,Research Article - Abstract
Background The role of tumor-associated macrophages (TAMs) in the cancer immune landscape and their potential as treatment targets or modulators of response to treatment are gaining increasing interest. TAMs display high molecular and functional complexity. Therefore their objective assessment as breast cancer biomarkers is critical. The aims of this study were to objectively determine the in situ expression and significance of TAM biomarkers (CD68, CD163, and MMP-9) in breast cancer and to identify subclasses of patients who could benefit from TAM-targeting therapies. Methods We measured CD68, CD163, and MMP-9 protein expression in formalin-fixed paraffin-embedded tissues of breast carcinomas represented in tissue microarray format using multiplexed quantitative immunofluorescence (QIF) in two independent Yale cohorts: cohort A—n = 398, estrogen receptor–positive (ER+) and ER− cases—and the triple-negative breast cancer (TNBC)-only cohort B (n = 160). Associations between macrophage markers, ER status, and survival were assessed. Protein expression measured by QIF was compared with mRNA expression data from the METABRIC study. Results All three macrophage markers were co-expressed, displaying higher expression in ER− cancers. High pan-macrophage marker CD68 correlated with poorer overall survival (OS) only in ER− cases of cohort A (P = 0.02). High expression of CD163 protein in TAMs was associated with improved OS in ER− cases (cohort A, P = 0.03 and TNBC cohort B, P = 0.04, respectively) but not in ER+ cancers. MMP-9 protein was not individually associated with OS. High expression of MMP-9 in the CD68+/CD163+ TAMs was associated with worse OS in ER+ tumors (P
- Published
- 2018
- Full Text
- View/download PDF
30. Immunological differences between primary and metastatic breast cancer
- Author
-
Giampaolo Bianchini, Meiling Zhang, Xiaotong Li, Tristen S. Park, Andrea Silber, Vikram B. Wali, Veerle Bossuyt, Vasiliki Pelekanou, Christos Hatzis, Malini Harigopal, Lajos Pusztai, Courtney Frederick, Borbála Székely, David L. Rimm, Qin Yan, and G Patwardhan
- Subjects
Adult ,0301 basic medicine ,Adolescent ,Biopsy ,medicine.medical_treatment ,Breast Neoplasms ,chemical and pharmacologic phenomena ,C-C chemokine receptor type 7 ,B7-H1 Antigen ,Metastasis ,Young Adult ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Mutation Rate ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,Cytotoxic T cell ,Medicine ,Lymphocyte Count ,Immunologic Surveillance ,Aged ,business.industry ,Cancer ,Hematology ,Immunotherapy ,Middle Aged ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,Gene Expression Regulation ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Female ,Tumor Escape ,business ,CD8 - Abstract
Background Little is known about how the immune microenvironment of breast cancer evolves during disease progression. Patients and methods We compared tumor infiltrating lymphocyte (TIL) count, programmed death-ligand 1 (PD-L1) protein expression by immunohistochemistry and mRNA levels of 730 immune-related genes using Nanostring technology in primary and metastatic cancer samples. Results TIL counts and PD-L1 positivity were significantly lower in metastases. Immune cell metagenes corresponding to CD8, T-helper, T-reg, Cytotoxic T, Dendritic and Mastoid cells, and expression of 13 of 29 immuno-oncology therapeutic targets in clinical development including PD1, PD-L1, and CTLA4 were significantly lower in metastases. There was also coordinated down regulation of chemoattractant ligand/receptor pairs (CCL19/CCR7, CXCL9/CXCR3, IL15/IL15R), interferon regulated genes (STAT1, IRF-1,-4,-7, IFI-27,-35), granzyme/granulysin, MHC class I and immune proteasome (PSMB-8,-9,-10) expression in metastases. Immunotherapy response predictive signatures were also lower. The expression of macrophage markers (CD163, CCL2/CCR2, CSF1/CSFR1, CXCR4/CXCL12), protumorigenic toll-like receptor pathway genes (CD14/TLR-1,-2,-4,-5,-6/MyD88), HLA-E, ecto-nuclease CD73/NT5E and inhibitory complement receptors (CD-59,-55,-46) remained high in metastases and represent potential therapeutic targets. Conclusions Metastatic breast cancers are immunologically more inert than the corresponding primary tumors but some immune-oncology targets and macrophage and angiogenesis signatures show preserved expression and suggest therapeutic combinations for clinical testing.
- Published
- 2018
- Full Text
- View/download PDF
31. Abstract P2-09-06: Quantitative spatial profiling of tumor associated macrophages and the PD-1/PD-L1 interaction in breast cancer
- Author
-
Vasiliki Pelekanou, Lajos Pusztai, David L. Rimm, and Veronique Neumeister
- Subjects
Oncology ,Cancer Research ,Tumor microenvironment ,medicine.medical_specialty ,Tissue microarray ,biology ,business.industry ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Transcriptome ,Breast cancer ,Internal medicine ,PD-L1 ,medicine ,biology.protein ,Biomarker (medicine) ,business - Abstract
Background: Although immunotherapy approaches are being successfully administered in some breast cancer (BC) patients (pts), biomarkers of response remain elusive. Tumor associated macrophages (TAMs) are the most prominent immune cells in breast tumors, mediating the cross-talk between tumor cells and tumor infiltrating lymphocytes (TILs). Specific biomarkers of breast TAMs' functional status remain to be defined. CSF-1R, is a TAMs regulator and key target across cancers clinically tested, alone or combined with anti-PD-1 checkpoint inhibitors. The goals of the study were: 1) to objectively measure CSF-1R expression within all CD68+ and M2-like CD163+TAMs, as well as PD-L1/PD-1 spatial interaction and 2) to determine whether objectively quantifying these key immune mechanisms related to TAMs immunomodulation within the tumor microenvironment can predict outcome and potentially response to immunotherapy. Methods: Tissue Microarrays (TMAs) from two Yale BC cohorts (Cohort A, all breast cases, n=320) (Cohort B, TNBC, n=132) were assessed by quantitative immunofluorescence (QIF) for CSF-1R/CD163/CD68; PD-1/PD-L1 interaction score (proportion of PD-1+ cells co-localized with PD-L1) and co-expression of the multiplexed biomarker panels. Biomarker positive cells and their co-localization were objectively measured using the AQUA method of QIF. QIF scores were compared by linear regression coefficients (R2). Overall and recurrence-free survival (OS and RFS) were assessed. Our protein data were compared with transcriptome data from the METABRIC study obtained from www.cbioportal.org. Results: CSF-1R expression was associated with expression of both CD68 and CD163 in both cohorts (A: R2=0.64, B: R2=0.49). CSF-1R in CD163/CD68 was higher in TNBC Cohort B (p PD-L1 mostly co-localized with CD68 TAMs (R2=0.7). Tumor PD-L1 tended to be mutually exclusive of CSF-1R. PD-L1/PD-1 colocalization was higher in TNBC (p The trend of mutual exclusivity between CSF-1R in TAMs and PD-1/PD-L1 was confirmed by expression (mRNA) data from METABRIC study. Discussion: This novel multiplexed method profiling key tumor-immune suppression pathways could identify BC pts likely to respond to anti-PD-1/anti-CSF-1R therapy. This method could help stratify pts for mono- or combined therapy in future clinical trials. Citation Format: Pelekanou V, Neumeister V, Pusztai L, Rimm DL. Quantitative spatial profiling of tumor associated macrophages and the PD-1/PD-L1 interaction in breast cancer [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P2-09-06.
- Published
- 2018
- Full Text
- View/download PDF
32. Identification and validation of a novel biologics target in triple negative breast cancer
- Author
-
Thomas Karn, Vikram B. Wali, Lajos Pusztai, Bryce Nelson, Vasiliki Pelekanou, Qin Yan, Alberto Ocaña, Jian Cao, Christos Hatzis, and Gauri A. Patwardhan
- Subjects
Immunoconjugates ,Cell ,Datasets as Topic ,lcsh:Medicine ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Mertansine ,Article ,Immunoglobulin Fab Fragments ,Mice ,chemistry.chemical_compound ,Breast cancer ,Drug Development ,In vivo ,Target identification ,Cell Line, Tumor ,Gene expression ,medicine ,Animals ,Humans ,Maytansine ,Breast ,GABA-A Receptor Antagonists ,Molecular Targeted Therapy ,ddc:610 ,lcsh:Science ,Triple-negative breast cancer ,Cell Proliferation ,Gene knockdown ,Multidisciplinary ,Gene Expression Profiling ,Cell Membrane ,lcsh:R ,Receptors, GABA-A ,Xenograft Model Antitumor Assays ,Gene expression profiling ,medicine.anatomical_structure ,chemistry ,Cell culture ,Gene Knockdown Techniques ,Cancer research ,Female ,lcsh:Q - Abstract
The goal of this study was to identify a novel target for antibody-drug conjugate (ADC) development in triple negative breast cancer (TNBC), which has limited treatment options, using gene expression datasets and in vitro siRNA/CRISPR and in vivo functional assays. We analyzed 4467 breast cancers and identified GABRP as top expressed gene in TNBC with low expression in most normal tissues. GABRP protein was localized to cell membrane with broad range of receptors/cell (815–53,714) and expressed by nearly half of breast cancers tissues. GABRP gene knockdown inhibited TNBC cell growth and colony formation in vitro and growth of MDA-MB-468 xenografts in nude mice. Commercially available anti-GABRP antibody (5–100 μg/ml) or de novo generated Fabs (20 μg/ml) inhibited TNBC cell growth in vitro. The same antibody conjugated to mertansine (DM1) also showed significant anticancer activity at nanomolar concentrations. Our results indicate that GABRP is a potential novel therapeutic target for ADC development.
- Published
- 2019
33. Immune profiling of pre- and post-treatment breast cancer tissues from the SWOG S0800 neoadjuvant trial
- Author
-
Mingdong Liu, Xiaotong Li, Zeina Nahleh, Vasiliki Pelekanou, Sarah Warren, Daniel F. Hayes, Nathane Elliott, Patrick Danaher, Gabriel N. Hortobagyi, William E. Barlow, Vikram B. Wali, Christos Hatzis, Lajos Pusztai, and Alessandra Cesano
- Subjects
PD-L1 ,0301 basic medicine ,Neoadjuvant treatment ,Cancer Research ,Myeloid ,medicine.medical_treatment ,Immunology ,Breast Neoplasms ,chemical and pharmacologic phenomena ,lcsh:RC254-282 ,B7-H1 Antigen ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Pharmacology ,Tumor microenvironment ,PDGFB ,biology ,Tumor-infiltrating lymphocytes ,business.industry ,Gene Expression Profiling ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune-related genes ,Neoadjuvant Therapy ,Tumor infiltrating lymphocytes ,3. Good health ,Bevacizumab ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,Female ,business ,Research Article - Abstract
Background How the immune microenvironment changes during neoadjuvant chemotherapy of primary breast cancer is not well understood. Methods We analyzed pre- and post-treatment samples from 60 patients using the NanoString PanCancer IO360™ assay to measure the expression of 750 immune-related genes corresponding to 14 immune cell types and various immune functions, and assessed TIL counts and PD-L1 protein expression by immunohistochemistry. Treatment associated changes in gene expression levels were compared using t-test with Bonferroni correction. TIL count, PD-L1 protein and immune metagenes were compared using Wilcoxon test. Baseline immune markers were correlated with pathologic complete response (pCR) using estrogen receptor and treatment arm adjusted logistic regression. Results At baseline, high TIL counts and high expression of chemoattractant cytokines (CCL21, CCL19) and cytotoxic T cell markers were associated with higher pCR rate. High expression of stromal genes (VEGFB, TGFB3, PDGFB, FGFR1, IGFR1), mast and myeloid inflammatory cell metagenes, stem cell related genes (CD90, WNT11, CTNNB1) and CX3CR1, and IL11RA were associated with residual disease (RD). After treatment, in cases with pCR, TIL counts and most immune genes decreased significantly. Among RD cases, TIL counts and PD-L1 expression did not change but cellular stress and hypoxia associated genes (DUSP1, EGR1), and IL6, CD36, CXCL2, CD69 and the IL8/VEGF metagene increased. Conclusions Activated T cells in the tumor microenvironment are associated with pCR whereas stromal functions are associated with residual disease. Most immune functions decrease during neoadjuvant chemotherapy but several immunotherapy targets (PD-L1, IL6, IL8) remain expressed in RD suggesting potential therapeutic strategies. Electronic supplementary material The online version of this article (10.1186/s40425-019-0563-7) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
34. Abstract 406: Development of a gene signature assessing ER modulation by SERMs and SERDs as a target engagement biomarker for endocrine therapy in breast cancer
- Author
-
Emma Wang, Alice Gosselin, Alexei Protopopov, Colette Dib, Joon Sang Lee, Maysoun Shomali, Vasiliki Pelekanou, Nils Ternes, Christopher Soria, Patrick Cohen, Marina Celanovic, Monsif Bouaboula, Wilson Dos-Santos-Bele, Jack Pollard, and Eric Boitier
- Subjects
Cancer Research ,Fulvestrant ,medicine.drug_class ,business.industry ,Cancer ,Estrogen receptor ,Gene signature ,medicine.disease ,Metastatic breast cancer ,Breast cancer ,Oncology ,Estrogen ,Selective estrogen receptor modulator ,medicine ,Cancer research ,business ,medicine.drug - Abstract
Hormone or endocrine therapy is the primary treatment for estrogen receptor-positive (ER+) breast cancer. Selective Estrogen Receptor Modulators (SERMs) have been used for women with ER+ invasive cancer in the adjuvant setting. However, they do not fully inhibit ER transcriptional activity. Therefore, selective estrogen receptor degraders (SERDs) such as fulvestrant have been developed to overcome the partial modulation of ER transcriptional activity by SERMs. However, the clinical benefit of fulvestrant is limited by its pharmaceutical properties, and burden of intramuscular administration. We developed amcenestrant (SAR439859), a novel, orally bioavailable SERD to improve drug properties and is under clinical investigation.To assess the relative ER modulating activity, we developed a gene signature by transcriptional profiling of multiple ER+ cell lines. We identified 87 genes that were either up- or down-regulated by estradiol and reversed by SERM and SERD compounds or differentially expressed between a SERM/SERD compound and estradiol. We assessed ER activity scores by applying Gene Set Variation Analysis (GSVA) method to the ER gene signature and evaluated the effect of 6 SERD compounds. Amcenestrant and fulvestrant illuminated a deeper inhibition of ER activity compared to the other compounds. In addition, these ER activity scores could potentially be utilized in clinics as pharmacodynamic (PD) biomarker to assess target engagement. We applied this ER signature to RNA-seq data from paired pre- and post-treatment tumor biopsies from patients of NCT03284957 study (AMEERA-1), an open-label, phase 1/2 study of amcenestrant in postmenopausal women with ER+/HER2- metastatic breast cancer. The results of our analysis showed that ER activity is down-regulated by amcenestrant (oral SERD) for 3 out of 5 patients, in parallel with clinical benefit. Validation in a larger patient cohort is needed. Citation Format: Joon Sang Lee, Maysoun Shomali, Monsif Bouaboula, Emma Wang, Wilson Dos-Santos-Bele, Colette Dib, Eric Boitier, Vasiliki Pelekanou, Nils Ternes, Alice Gosselin, Patrick Cohen, Marina Celanovic, Christopher Soria, Alexei Protopopov, Jack Pollard. Development of a gene signature assessing ER modulation by SERMs and SERDs as a target engagement biomarker for endocrine therapy in breast cancer [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 406.
- Published
- 2021
- Full Text
- View/download PDF
35. Abstract 739: Preclinical and clinical activity of SAR439859, Amcenestrant, a next generation SERD
- Author
-
Fangxian Sun, Youssef El-Ahmed, Monsif Bouaboula, Vasiliki Pelekanou, Marina Celanovic, Laurent Debussche, Christina I. Herold, Chris Soria, Joon Sang Lee, Jack Pollard, Patrick Cohen, Maysoun Shomali, Laurent Schio, Laurent Besret, and Anne Caron
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Internal medicine ,medicine ,Bioinformatics ,business - Abstract
Primary treatment for estrogen receptor-positive (ER+) breast cancer is endocrine therapy. Selective estrogen receptor degraders (SERDs), such as fulvestrant, induce effective ER signaling inhibition, although clinical studies with fulvestrant report insufficient blockade of ER signaling, possibly due to suboptimal pharmaceutical properties. Here we describe the discovery of SAR439859 (Amcenestrant), a novel, orally bioavailable SERD with potent antagonist and degradation activities against both wild-type and mutant Y537S ER. Driven by its fluoropropyl pyrrolidinyl side chain, SAR439859 has demonstrated broader and superior ER antagonist and degrader activities across a large panel of ER+ cells, compared with other SERDs including improved inhibition of ER signaling and tumor cell growth. Similarly, in vivo treatment with SAR439859 demonstrated significant tumor regression in ER+ breast cancer models including MCF7-ESR1 mutant-Y537S mouse tumors and HCI013, a patient-derived tamoxifen-resistant xenograft tumor. Importantly, in these mutant ESR1 models, we demonstrate that the efficacy of SAR439859 was increased when coadministered with palbociclib, an inhibitor of cyclin-dependent kinase 4 and 6. In the clinical setting, SAR439859 also demonstrated a high level of target engagement, ER degradation and inhibition of ER signaling as well as encouraging anti-tumor activity in heavily pretreated patients with advanced or metastatic ER-positive breast cancer. These findings indicate that SAR439859 would provide therapeutic benefit to patients with ER+ breast cancer, including those who are resistant to endocrine therapy with both wild-type and mutant ER. Citation Format: Maysoun Shomali, Fangxian Sun, Laurent Besret, Anne Caron, Joon Sang Lee, Youssef El-Ahmed, Laurent Schio, Jack Pollard, Vasiliki Pelekanou, Patrick Cohen, Marina Celanovic, Christina Herold, Chris Soria, Laurent Debussche, Monsif Bouaboula. Preclinical and clinical activity of SAR439859, Amcenestrant, a next generation SERD [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 739.
- Published
- 2021
- Full Text
- View/download PDF
36. Abstract 1825: SAR443216, a novel trispecific T cell engager with potent T cell-dependent cytotoxicity for HER2-low tumors
- Author
-
Zhi Yong Yang, Lan Wu, Zhen Xing, Ronnie Wei, Sri Vadde, Serena Masciari, Liqing Chen, Michele Sanicola-Nadel, Sukhvinder Sidhu, Virna Cortez-Retamozo, Lily Pao, Zhili Song, Dmitri Wiederschain, Vasiliki Pelekanou, Wenwen Sha, Dinesh S. Bangari, Edward Seung, and Gary J. Nabel
- Subjects
Cancer Research ,medicine.anatomical_structure ,Oncology ,Chemistry ,T cell ,Cancer research ,medicine ,skin and connective tissue diseases ,Cytotoxicity - Abstract
Current HER2-targeted therapies have markedly improved the outcome of cancer patients with HER2 overexpressing tumors. However, these patients may eventually relapse or develop treatment resistance. In addition, HER2-low patients that are not eligible for treatment constitute a significant portion of the breast cancer patients. To address these unmet needs, we have developed a novel HER2-targeting T cell engager, SAR443216. This is a trispecific antibody with binding sites for HER2, CD3 and CD28, and containing a mutated IgG4-Fc which lacks effector functions. CD28 binding contributes to T cell activation, including activation of IL-2 and NFκB pathways, as well as induction of anti-apoptotic protein, Bcl-xL. In the presence of HER2-positive cancer cells, SAR443216 is able to activate primary human CD4 and CD8 T cells, resulting in T cell proliferation and secretion of cytokines and granzyme B. Moreover, it has potent in vitro T cell-dependent cellular cytotoxicity (TDCC) against a panel of HER2-expressing cancer cell lines, including those that are HER2-low. The potency of in vitro TDCC is largely correlated with HER2 surface expression in the target cells. Finally, in a HER2-low breast cancer xenograft model, SAR443216 also exhibited significant anti-tumor activity in immuno-deficient NSG mice reconstituted with primary human T cells. Thus, SAR443216 represents a promising new drug for cancer patients with HER2-expressing tumors, including those who are currently ineligible for stand-of-care therapy. Citation Format: Wenwen Sha, Sri Vadde, Zhili Song, Edward Seung, Zhen Xing, Liqing Chen, Virna Cortez-Retamozo, Sukhvinder Sidhu, Dinesh Bangari, Lan Wu, Ronnie Wei, Zhi-yong Yang, Gary Nabel, Vasiliki Pelekanou, Michele Sanicola-Nadel, Serena Masciari, Dmitri Wiederschain, Lily Pao. SAR443216, a novel trispecific T cell engager with potent T cell-dependent cytotoxicity for HER2-low tumors [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 1825.
- Published
- 2021
- Full Text
- View/download PDF
37. Characterization of the tumor immune microenvironment of triple-negative breast cancer (TNBC) patients who self-identify as African American (AA) or non-African American (NonAA)
- Author
-
Yalai Bai, Tao Qing, Xiaotong Li, David L. Rimm, Kim Blenman, Kimberly Cole, Vesal Yaghoobi, Vasiliki Pelekanou, Emily Reisenbichler, Lajos Pusztai, Michal Marczyk, Eiman Y Ibrahim, Vignesh Gunasekharan, and Tess O'Meara
- Subjects
Oncology ,African american ,Cancer Research ,medicine.medical_specialty ,business.industry ,Immune microenvironment ,Internal medicine ,Incidence (epidemiology) ,medicine ,business ,Triple-negative breast cancer - Abstract
564 Background: What tumor biological differences, if any, contribute to the higher incidence and worse prognosis of triple negative breast cancer (TNBC) in African American (AA) compared to NonAA patients are unknown. We hypothesized that differences in the tumor immune microenvironment may contribute to the outcome disparities. The purpose of this study was to characterize and compare the immune microenvironment of TNBC between patients self-identified as NonAA or AA. Methods: Formalin fixed paraffin embedded surgically resected cancer and paired normal tissues collected before any systemic therapy and the corresponding clinical data were collected for NonAA (n = 56) and AA (n = 54) stage I-III TNBC treated at Yale Cancer Center between 2000-2017. The two cohorts were matched for clinical stage, age of diagnosis, and year of diagnosis. We performed somatic and germline whole exome sequencing (WES), bulk RNA sequencing, and immunohistochemistry to assess PD-L1 expression (SP142). Stromal tumor infiltrating lymphocytes (sTILs) were assessed on H&E slides. Mutation load, mutation frequencies, and gene expression differences were compared at gene and pathway level. Immune cell composition was estimated through gene expression deconvolution analyses (TIDE). Results: Tumor mutational burden was similar between the two cohorts. At gene level, few genes had significantly different somatic mutation frequencies, or differential mRNA expression between AA and NonAA samples. Pathway level alterations showed inflammation, immunity (adaptive; innate), antigen presentation, and allograft rejection pathways were more affected by somatic mutations in AA samples. The affected genes differed from cancer to cancer and were not recurrent and therefore were missed at gene level analysis. Gene set enrichment and co-expression analysis also showed higher immune related pathway expression in AA samples. Unsupervised co-expression cluster analysis confirmed coordinated overexpression of genes involved in immunity, inflammation, and cytokine/chemokine signaling in AA patients. Two immunotherapy response predictive signatures, immune inflamed and the IFNG as well as sTILs score and PD-L1 positivity were also higher in AA samples. These findings raise the possibility that immune checkpoint inhibitors might be particularly effective in AA patients. In NonAA samples, the EMT transition, angiogenesis, adipogenesis, myogenesis, fatty acid metabolism, TGFβ signaling, UV-response, and hypoxia pathways were overexpressed. TIDE analysis suggested higher levels of TAM M2, overall TIDE score, and the Immune Exclusion score in NonAA samples. Conclusions: TNBC in AA patients more frequently harbor somatic mutations in genes involved with immune functions and overexpress immune and inflammatory genes compared to NonAA patients.
- Published
- 2021
- Full Text
- View/download PDF
38. AMEERA-5: A randomized, double-blind phase III study of amcenestrant (SAR439859) + palbociclib versus letrozole + palbociclib for previously untreated ER+/HER2- advanced breast cancer
- Author
-
Aditya Bardia, Sylvaine Cartot-Cotton, Joyce O'Shaughnessy, Shao Zhi-min, Vasiliki Pelekanou, Suzette Delaloge, Qianying Liu, Hiroji Iwata, D. Kanagavel, Javier Cortes, Patrick Cohen, and Sara A. Hurvitz
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Advanced breast ,Letrozole ,Cancer ,Estrogen receptor ,Palbociclib ,medicine.disease ,Double blind ,Internal medicine ,Medicine ,business ,medicine.drug - Abstract
TPS1104 Background: Selective estrogen receptor degraders (SERDs) block estrogen receptor (ER) associated signaling and have created interest for treating patients (pts) with advanced ER+ breast cancer (BC). Fulvestrant is currently the only SERD available for advanced BC but requires intramuscular administration, limiting the applied dose, exposure and receptor engagement. Amcenestrant (SAR439859) is an oral SERD that binds with high affinity to both wild-type and mutant ER, blocking estradiol binding and promoting up to 98% ER degradation in preclinical studies. In the phase I AMEERA-1 study of pretreated pts with ER+/HER2- advanced BC, amcenestrant 150–600 mg once daily (QD) showed a mean ER occupancy of 94% with plasma concentrations > 100 ng/mL and a favorable safety profile (Bardia, 2019; data on file). Combination therapy with amcenestrant + palbociclib (palbo) was also evaluated as part of this ongoing phase I study. CDK 4/6 inhibitors (CDK4/6i) combined with an aromatase inhibitor (AI), the gold standard for first line treatment for advanced breast cancer, prolong progression free survival (PFS) in pts with no prior treatment for ER+/HER2- advanced BC, but OS benefit has not been shown yet in postmenopausal pts. There remains a clinical need for more effective treatments in this setting. Methods: AMEERA-5 (NCT04478266) is an ongoing, prospective, randomized, double-blind phase III study comparing the efficacy and safety of amcenestrant + palbo with that of letrozole + palbo in pts with advanced, locoregional recurrent or metastatic ER+/HER2- BC who have not received prior systemic therapy for advanced disease. The study includes men, pre/peri-menopausal (with goserelin) and post-menopausal women. Pts with progression during or within 12 months of (neo)adjuvant endocrine therapy using any of the following agents are excluded: AI, selective estrogen receptor modulators, CDK4/6i. Pts are randomized 1:1 to either continuous amcenestrant 200 mg or letrozole 2.5 mg QD orally with matching placebos; both combined with palbo 125 mg QD orally (d1–21 every 28-d cycle). Randomization is stratified according to disease type (de novo metastatic vs recurrent disease), the presence of visceral metastasis, and menopausal status. The primary endpoint is investigator assessed progression free survival (PFS) (RECIST v1.1). Secondary endpoints are overall survival, PFS2, objective response rate, duration of response, clinical benefit rate, pharmacokinetics of amcenestrant and palbo, health-related quality of life, time to chemotherapy, and safety. Biomarkers will be measured in paired tumor biopsies and cell free deoxyribonucleic acid (cfDNA) over time. Target enrolment = 1066 pts; enrolment as of 1/2021 = 33 pts. Bardia A, et al., J Clin Oncol. 2019; 37 (15 suppl):1054 Clinical trial information: NCT04478266 .
- Published
- 2021
- Full Text
- View/download PDF
39. Abstract PD8-08: A phase 1/2 study of SAR439859, an oral selective estrogen receptor (ER) degrader (SERD), as monotherapy and in combination with other anti-cancer therapies in postmenopausal women with ER-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC): AMEERA-1
- Author
-
Marina Celanovic, Mario Campone, Alice Gosselin, Sarat Chandarlapaty, Aditya Bardia, Séverine Doroumian, Hannah M. Linden, Vasiliki Pelekanou, and Gary A. Ulaner
- Subjects
Cancer Research ,Postmenopausal women ,Oncology ,business.industry ,Cancer research ,medicine ,Estrogen receptor ,Cancer ,medicine.disease ,business ,Human Epidermal Growth Factor Receptor 2 ,Metastatic breast cancer - Abstract
Background SERDs belong to a class of ER-targeted therapies that antagonize and degrade ERs, including in ER-dependent tumors resistant to other endocrine therapies (ET). This study (AMEERA-1; NCT03284957) investigates SAR439859, an oral SERD, as monotherapy and (in ongoing cohorts) in combination with targeted therapies in patients (pts) with ER+/HER2- mBC. Here we report updated safety and antitumor activity with SAR439859 monotherapy, including exploratory analyses by prior therapy and ESR1 status. Methods This open-label, phase 1/2, first-in-human study assessed SAR439859 as monotherapy in Parts A (dose escalation 20-600 mg once daily [QD]) and B (dose expansion with recommended dose at 400 mg QD). Eligible pts were heavily pre-treated, postmenopausal women with ER+/HER2- mBC and measurable disease who received ≥6 months of prior ET in the advanced setting. Prior chemotherapy, mammalian target of rapamycin inhibitors (mTORi) and cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) for advanced disease were allowed. This analysis pooled data from pts receiving SAR439859 ≥150 mg (Part A) and 400 mg (Part B), administered in 28-day cycles. Antitumor activity was assessed by the objective response rate and clinical benefit rate (CBR: complete response [CR], partial response [PR] and stable disease [SD] ≥24 weeks) per RECIST v1.1, determined by investigators. Analyses by prior therapy and baseline ESR1 mutation status were performed. Safety was also evaluated. Results Pts (n = 62; Part A: 13; Part B: 49) had a median age of 63 (range 37-88) years and ECOG PS 0 (59.7%) or 1 (40.3%); 93.5% had visceral metastases. Pts had a median of 2 (range 1-8) prior lines of therapy in the advanced setting (48.4% had ≥3 prior lines): all had prior ET and 72.6% had prior targeted therapy. SAR439859 monotherapy showed antitumor activity in the response-evaluable population (n = 59) and in subset populations with ≤3 prior lines (n = 33) or without prior mTORi, CDK4/6i, or SERD (n = 14) (Table 1). For pts with ESR1 status (n = 58), CBR with SAR439859 was comparable in ESR1 wild-type (36.7%) and mutant mBC (32.1%), with similar results in subpopulations. Treatment-related adverse events (TRAEs) occurred in 62.9% of pts (all grade 1-2); none resulted in SAR439859 discontinuation. Most frequent TRAEs were hot flush (16.1%); constipation and arthralgia (each 9.7%); decreased appetite, vomiting, diarrhea and nausea (each 8.1%); and fatigue (6.5%). Conclusions Among heavily pre-treated pts, SAR439859 demonstrated antitumor activity, similar to historical single-agent fulvestrant activity in less heavily pre-treated pts with advanced/mBC (2L+ setting; no prior targeted agents) (indirect comparison). In both subsets of pts with fewer prior advanced lines of therapy, SAR439859 showed trends of greater clinical activity versus historical fulvestrant activity. SAR439859 had a favorable safety profile with limited TRAEs. No safety signals of cardiac or ocular toxicities were observed. Ongoing parts of the study are investigating SAR439859 in combination with targeted therapies. Based on the monotherapy results, a randomized phase 2 study is investigating SAR439859 compared with physician’s choice in a 2L+ setting (AMEERA-3; NCT04059484). Funding: Sanofi. Antitumor activity overall and in subpopulations by prior lines of therapy (Parts A+B)Overall population (A+B)≤3 Prior advanced linesWithout prior targeted therapy(n = 59)a(n = 33)b(n = 14)cBOR, n (%)–CR000–PR5 (8.5)5 (15.2)3 (21.4)–SD24 (40.7)15 (45.5)8 (57.1)–PD30 (50.8)13 (39.4)3 (21.4)ORR, n (%)5 (8.5)5 (15.2)3 (21.4)CBR, n (%)20 (33.9)14 (42.4)9 (64.3)aPooled cohort (A ≥150 mg QD + B);bSubset of pooled cohort with ≤3 prior lines in the metastatic setting, including ≤1 of either prior chemotherapy or CDK4/6i and no prior mTORi; cSubset of pooled cohort with no prior mTORi, CDK4/6i, or fulvestrant.BOR, best overall response; CBR, clinical benefit rate; CR, complete response; ORR, objective response rate; PD, progressive disease; PR, partial response; SD, stable disease. Citation Format: Hannah M Linden, Mario Campone, Aditya Bardia, Gary A Ulaner, Alice Gosselin, Séverine Doroumian, Vasiliki Pelekanou, Marina Celanovic, Sarat Chandarlapaty. A phase 1/2 study of SAR439859, an oral selective estrogen receptor (ER) degrader (SERD), as monotherapy and in combination with other anti-cancer therapies in postmenopausal women with ER-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC): AMEERA-1 [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD8-08.
- Published
- 2021
- Full Text
- View/download PDF
40. Abstract OT-09-11: AMEERA-4, a phase 2 window study of SAR439859 vs letrozole in post-menopausal women with newly diagnosed estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer
- Author
-
Sylvaine Cartot-Cotton, Vasiliki Pelekanou, Christina I. Herold, Mario Campone, Qiuyan Wang, and Bethany Ling
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Aromatase inhibitor ,business.industry ,medicine.drug_class ,Letrozole ,Estrogen receptor ,Cancer ,medicine.disease ,Metastatic breast cancer ,law.invention ,Breast cancer ,Randomized controlled trial ,law ,Estrogen ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Background Endocrine therapy targeting estrogen receptor (ER) signaling is the standard of care for women with ER+ breast cancer. Selective ER degraders (SERDs) block ER signaling through dual competitive antagonism and receptor degradation. SAR439859, a potent, oral SERD, is in clinical development for ER+/HER- breast cancer. AMEERA-4 (NCT04191382; ACT16106) is a 14-day preoperative, non-therapeutic ‘window of opportunity’ trial to assess the direct effects of SAR439859 on tumor cell proliferation by evaluating the pharmacodynamic activity of SAR439859 in ER+/HER2- breast cancer. Methods This international, open-label, Phase 2 randomized study evaluates SAR439859 at two dose levels vs the aromatase inhibitor letrozole by assigning 126 preoperative patients 1:1:1 to receive SAR439859 400 mg/day, SAR439859 200 mg/day or letrozole 2.5 mg/day. SAR439859 dosing is based on an ongoing AMEERA-1 Phase 1/2 study (NCT03284957; TED14856) in metastatic breast cancer. Postmenopausal women with ER+/HER2- breast cancer indicated for immediate surgery (Stage I, Stage II or operable Stage III), Eastern Cooperative Oncology Group performance status 0-1, and Ki67 levels of ≥ 15% are eligible. Exclusion criteria include disorders potentially affecting absorption of SAR439859 or letrozole, and any prior therapy for breast cancer. Patients receive study treatment for 14 days, with the last dose given on the day before surgery. Paired tumor biopsies for assessment of biomarkers are performed at baseline and during surgery. The primary study endpoint is change in Ki67, a predictor of treatment benefit and long-term survival outcomes, after 14 days of treatment compared with baseline. Secondary endpoints include proportion of patients with ≥ 50% decrease in Ki67, ER expression to assess degradation, and safety. Pharmacokinetics of SAR439859, additional tumor markers, genomic mutation profile, Preoperative Endocrine Prognostic Index and pathological complete response will also be assessed. The study is currently recruiting. Funding: Sanofi. © 2020 American Society of Clinical Oncology, Inc. Reused with permission. This abstract was accepted and previously presented at the 2020 ASCO Annual Meeting. All rights reserved. Citation Format: Mario Campone, Christina Herold, Qiuyan Wang, Vasiliki Pelekanou, Sylvaine Cartot-Cotton, Bethany Ling. AMEERA-4, a phase 2 window study of SAR439859 vs letrozole in post-menopausal women with newly diagnosed estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr OT-09-11.
- Published
- 2021
- Full Text
- View/download PDF
41. Abstract OT-09-09: AMEERA-3, a phase 2 trial of SAR439859 vs endocrine monotherapy in pre- and post-menopausal, estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (her2−), locally advanced or metastatic breast cancer (BC) with prior exposure to hormonal therapies
- Author
-
Katharine Cuff, Sara M. Tolaney, Suzette Delaloge, Amele Amrate, Qianying Liu, Diego Kaen, Irfan Cicin, Vasiliki Pelekanou, Sylvaine Cartot-Cotton, Peter A. Kaufman, Rafael Betancourt, and Arlene Chan
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Aromatase inhibitor ,Fulvestrant ,business.industry ,medicine.drug_class ,Cancer ,Estrogen receptor ,medicine.disease ,Metastatic breast cancer ,Breast cancer ,Response Evaluation Criteria in Solid Tumors ,Internal medicine ,medicine ,business ,Tamoxifen ,medicine.drug - Abstract
Background Endocrine therapy (ET) targeting ER signaling is the mainstay of care for ER+ metastatic BC. There remains an unmet need in patients whose tumors become resistant to currently available ET. Selective ER degraders (SERDs) were developed to overcome resistance to existing ER-directed therapies by both competitively antagonizing and degrading ERs, while exploiting continued dependence of the tumor on ER signaling. SAR439859 is a potent SERD with robust preclinical ER degrading activity. In AMEERA-1, a Phase 1 dose escalation trial, SAR439859, had a favorable safety profile with no dose-limiting toxicities across all doses (20-600 mg once per day; QD). ER occupancy generally exceeded > 87% with plasma concentrations > 100 ng/mL. Overall response rate was 6.3% and the recommended Phase 2 monotherapy dose was 400 mg QD. Methods AMEERA-3 is an international, prospective, open-label, randomized Phase 2 study (NCT04059484; ACT16105) designed to assess safety and efficacy of SAR439859 in pts with ER+ (>1%)/HER2− metastatic or locally advanced BC progressing on ≥ 6 months of continuous ET (0-2 lines in the metastatic setting). Prior cyclin-dependent kinase (CDK) inhibitors are allowed. Exclusion criteria include: Eastern Cooperative Oncology Group performance status (ECOG PS) ≥ 2, life expectancy < 3 months, > 1 chemotherapy or targeted therapy in the metastatic setting, concomitant illness and factors potentially affecting SAR439859 absorption. Patients are randomized 1:1 to SAR439859 400 mg QD orally or physician’s choice of endocrine monotherapy (fulvestrant, tamoxifen, aromatase inhibitor). Patients receive 28-day cycles until unacceptable toxicity, progression, death, investigator decision, or patient request. Stratification factors include visceral metastases, prior CDK4/6 inhibitors, and ECOG PS. The primary endpoint is progression-free survival (Response Evaluation Criteria in Solid Tumors v1.1). Secondary endpoints include overall survival, response rate, duration of response, clinical benefit, pharmacokinetics, quality of life and safety. Target enrollment: n = 282; current enrollment: n = 9. Funding: Sanofi. © 2020 American Society of Clinical Oncology, Inc. Reused with permission. This abstract was accepted and previously presented at the 2020 ASCO Annual Meeting. All rights reserved. Citation Format: Sara Tolaney, Irfan Cicin, Rafael Betancourt, Arlene Chan, Diego Kaen, Peter Kaufman, Suzette Delaloge, Qianying Liu, Sylvaine Cartot-Cotton, Amele Amrate, Vasiliki Pelekanou, Katharine Cuff. AMEERA-3, a phase 2 trial of SAR439859 vs endocrine monotherapy in pre- and post-menopausal, estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (her2−), locally advanced or metastatic breast cancer (BC) with prior exposure to hormonal therapies [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr OT-09-09.
- Published
- 2021
- Full Text
- View/download PDF
42. Non-malignant respiratory epithelial cells preferentially proliferate from resected non-small cell lung cancer specimens cultured under conditionally reprogrammed conditions
- Author
-
Uwe Rix, Kenneth E. Huffman, John D. Minna, Eric B. Haura, Tingting Jiang, Oliver A. Hampton, Jamie K. Teer, Boning Gao, Vasiliki Pelekanou, Harsha Doddapaneni, Fumi Kinose, Joy Jayaseelan, Robert C. Doebele, Jennifer R. Peters-Hall, Kemp H. Kernstine, Wei Zhang, Luc Girard, Yuval Kluger, Dara L. Aisner, Melissa Coquelin, David A. Wheeler, Chunxian Huang, Marileila Varella-Garcia, Kyle R. Covington, Dwight H Oliver, Anh T. Le, David L. Rimm, Jerry W. Shay, Adi F. Gazdar, and Jianhong Hu
- Subjects
Male ,0301 basic medicine ,Gerontology ,Oncology ,Lung Neoplasms ,DNA Mutational Analysis ,Non malignant ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Tumor Cells, Cultured ,Cells, Cultured ,Aged, 80 and over ,rock inhibitor ,Middle Aged ,Primary tumor ,conditionally reprogrammed cells ,humanities ,3. Good health ,030220 oncology & carcinogenesis ,Female ,Non small cell ,Research Paper ,Adult ,medicine.medical_specialty ,DNA Copy Number Variations ,Respiratory Mucosa ,03 medical and health sciences ,Cell Line, Tumor ,Thoracic Oncology ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Lung cancer ,non-small cell lung cancer ,Aged ,Cell Proliferation ,cell culture ,Base Sequence ,business.industry ,Gene Expression Profiling ,Cancer ,Epithelial Cells ,medicine.disease ,Coculture Techniques ,030104 developmental biology ,A549 Cells ,Mutation ,Cancer cell ,Mouse Fibroblast ,business ,respiratory epithelial cells - Abstract
// Boning Gao 1, 2, 3 , Chunxian Huang 1, 2 , Kemp Kernstine 4 , Vasiliki Pelekanou 5 , Yuval Kluger 5 , Tingting Jiang 6 , Jennifer R. Peters-Hall 7 , Melissa Coquelin 7 , Luc Girard 1, 2, 3 , Wei Zhang 1, 2 , Kenneth Huffman 1, 2 , Dwight Oliver 8 , Fumi Kinose 9 , Eric Haura 9 , Jamie K. Teer 10 , Uwe Rix 11 , Anh T. Le 12 , Dara L. Aisner 13 , Marileila Varella-Garcia 12, 13 , Robert C. Doebele 12 , Kyle R. Covington 14 , Oliver A. Hampton 14 , Harsha V. Doddapaneni 14 , Joy C. Jayaseelan 14 , Jianhong Hu 14 , David A. Wheeler 14 , Jerry W. Shay 7 , David L. Rimm 5 , Adi Gazdar 1, 2, 8 , John D. Minna 1, 2, 3, 15 1 Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA 2 The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA 3 Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA 4 Division of Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA 5 Department of Pathology, Yale University, New Haven, CT, USA 6 Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA 7 Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA 8 Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA 9 Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA 10 Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA 11 Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA 12 Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA 13 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA 14 Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA 15 Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA Correspondence to: Adi Gazdar, email: Adi.Gazdar@UTSouthwestern.edu John D. Minna, email: John.Minna@UTSouthwestern.edu Keywords: conditionally reprogrammed cells, respiratory epithelial cells, non-small cell lung cancer, rock inhibitor, cell culture Received: September 23, 2016 Accepted: December 20, 2016 Published: December 29, 2016 ABSTRACT The “conditionally reprogrammed cells” (CRC) method, using a Rho kinase inhibitor and irradiated mouse fibroblast cells has been described for the efficient growth of cells from malignant and non-malignant samples from primary tumor and non-malignant sites. Using the CRC method, four institutions independently cultured tumor tissues from 48 non-small cell lung cancers (NSCLC, mostly from primary resected tumors) and 22 non-malignant lungs. We found that epithelial cells could be cultured from tumor and non-malignant lung. However, epithelial cells cultured from tumors had features of non-malignant respiratory epithelial cells which include: 1) among 22 mutations found in the original tumors only two mutations were found in the CRC cultures with reduced frequency (31% to 13% and 92% to 15% from original tumor and CRC culture respectively); 2) copy number variation was analyzed in 9 tumor and their CRC cultures and only diploid patterns were found in CRC cultures; 3) mRNA expression profiles were similar to those of normal respiratory epithelial cells; and 4) co-culture of tumor and non-malignant lung epithelial cells resulted in mostly non-malignant cells. We conclude that CRC method is a highly selective and useful method for the growth of non-malignant respiratory epithelial cells from tumor specimens and only occasionally do such CRC cultures contain a small subpopulation of cancer cells marked by oncogenic mutations. While our findings are restricted to resected primary NSCLC, they indicated the necessity to fully characterize all CRC cultures and the need to develop culture technology that facilitates the growth of primary lung cancers.
- Published
- 2016
- Full Text
- View/download PDF
43. Androgen Control in Prostate Cancer
- Author
-
Elias Castanas and Vasiliki Pelekanou
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Prostatectomy ,medicine.medical_treatment ,Context (language use) ,Cell Biology ,Disease ,medicine.disease ,Biochemistry ,Management of prostate cancer ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Clinical research ,030220 oncology & carcinogenesis ,Internal medicine ,Localized disease ,medicine ,Biomarker (medicine) ,business ,Molecular Biology - Abstract
Research on prostate cancer has extensively advanced in the past decade, through an improved understanding for its genetic basis and risk-stratification. Molecular classification of prostate cancer into distinct subtypes and the recognition of new histologic entities promise the development of tailored-made management strategies of patients. Nowadays, various alternatives are available for clinical management of localized disease ranging from observation alone through radical prostatectomy. In patients with castration-resistant prostate cancer, the approval of new drugs for the management of metastatic disease has offered promising results improving the survival of these patients. In this context, androgen receptors (AR) remain at the epicenter of prostate cancer research holding a prominent role in the biology and therapeutic regimens of prostate cancer. As many of castration-resistant tumors retain hormone-responsiveness, AR is a clinical relevant, druggable target. However, AR paradoxically remains neglected as a prostate cancer biomarker. The great advancements in prostate cancer preclinical and clinical research, imply further improvement in clinical and translational data, for patient selection and treatment optimization. For a precision medicine-guided clinical management of prostate cancer, AR evaluation has to be implemented in companion and complementary diagnostics, as discussed here. J. Cell. Biochem. 117: 2224-2234, 2016. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
- Full Text
- View/download PDF
44. 277MO SAR439859, an oral selective estrogen receptor (ER) degrader (SERD), in ER+/ HER2- metastatic breast cancer (mBC): Biomarker analyses from a phase I/II study
- Author
-
Aditya Bardia, A-L. Bauchet, Vasiliki Pelekanou, Marina Celanovic, N. Ternes, Sarat Chandarlapaty, J. Sang Lee, V. Boutet, Mario Campone, A. Protopopov, W. Dos-Santos Bele, E. Boitier, Hannah M. Linden, J. Ming, A. Gosselin, and Simon Lord
- Subjects
Phase i ii ,Oncology ,business.industry ,medicine ,Cancer research ,Estrogen receptor ,Biomarker (medicine) ,Hematology ,medicine.disease ,business ,Metastatic breast cancer - Published
- 2020
- Full Text
- View/download PDF
45. Abstract 2001: Biomarker discovery in immunotherapy-treated melanoma patients with imaging mass cytometry
- Author
-
David L. Rimm, Harriet M. Kluger, Brian Bourke-Martin, Sandra Martinez-Morilla, Thazin New Aung, Franz Villarroel-Espindola, Pok Fai Wong, Maria I. Toki, and Vasiliki Pelekanou
- Subjects
0301 basic medicine ,CD20 ,Cancer Research ,Tumor microenvironment ,Tissue microarray ,biology ,business.industry ,Melanoma ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,Mass cytometry ,Biomarker discovery ,business - Abstract
Background: Understanding the complexity of the tumor microenvironment could allow identification of new biomarkers that predict response to immunotherapy in cancer patients and anticipate the toxicity associated with the treatment. Since the number of targets measurable by quantitative immunofluorescence (QIF) is limited, we applied Imaging Mass Cytometry (IMC), a metal-conjugated antibody-based high-plex technology, to generate a multidimensional analysis of the microenvironment. We interrogated a 25-antibody panel, including tumor and immune cell markers, in a cohort of melanoma patients treated with immune checkpoint blockers. Methods: We used a validated, optimized panel of 25 antibodies conjugated to unique metals, including a DNA intercalator conjugated with 191/193Ir, for detection using HyperionTM (Fluidigm). Digital image analysis was performed using a newly designed version of the AQUA software (Navigate BP) to measure marker intensity in molecularly defined masks or compartments as follows: all cells (DNA intercalator Ir191/193), tumor cells (HMB45/S100), stroma (tumor cells subtracted from all cells), T cells (CD3), B cells (CD20) and macrophages (CD68). Results were compared to QIF and Digital Spatial Profiling (DSP), a NanoString technology based on primary antibodies conjugated to indexing DNA oligos. We examined archived formalin-fixed paraffin-embedded (FFPE) samples from a metastatic melanoma cohort treated with immunotherapy (n=60) in tissue microarray format. Results: Immune markers such as CD3, CD4 and CD8 showed a high correlation between the three multiplexing methods: IMC, DSP and QIF (r=0.48-0.89, p Conclusion: Using IMC technology and digital image analysis based on pixel colocalization, we were able to evaluate simultaneously 25 markers on FFPE tissue microarray samples. There was a high concordance with QIF and DSP for immune markers such as CD3, CD4 and CD8. A series of potentially predictive biomarkers for immunotherapy in metastatic melanoma were identified. Citation Format: Sandra Martinez-Morilla, Franz Villarroel-Espindola, Pok Fai Wong, Harriet Kluger, Maria Toki, Thazin New Aung, Vasiliki Pelekanou, Brian Bourke-Martin, David L. Rimm. Biomarker discovery in immunotherapy-treated melanoma patients with imaging mass cytometry [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2001.
- Published
- 2020
- Full Text
- View/download PDF
46. Abstract 4973: Comparison of tumor immune microenvironment in triple negative breast cancer (TNBC) of African American versus that of white patients
- Author
-
Vasiliki Pelekanou, Andrea Silber, Kim Blenman, Vesal Yaghoobi, David L. Rimm, Lajos Pusztai, and Tess O'Meara
- Subjects
CD20 ,Cancer Research ,Tumor microenvironment ,education.field_of_study ,Stromal cell ,biology ,business.industry ,Lymphocyte ,Population ,Cancer ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Medicine ,business ,education ,CD8 ,Triple-negative breast cancer - Abstract
Introduction: Although there are many possible causes, it has been observed that Triple Negative Breast Cancer (TNBC) has a worse overall outcome in African American (AA) than white patients. Recently, there is also evidence that the patients response to the tumor, or the biological response expressed as the tumor immune microenvironment, impacts outcome. The goal of this study was to compare the Tumor Microenvironment (TME) of TNBC in the two mentioned groups in order to seek molecular parameters that are different in TNBC in AA patients as compared to whites. Methods: We selected N=50 AA and N=50 white TNBC samples from the Yale Pathology archives that were matched by diagnosis date. We measured CD45, CD3, CD8, CD20, CD14, CD68, PD-L1, THY1, aSMA and Fibroblast Activation Protein (FAP) expression in both the tumor and stromal compartments in whole slides from formalin fixed paraffin embedded (FFPE) tissues using multiplexed quantitative immunofluorescence (QIF). We also assessed the activation status of CD3-positive cells using Ki67 and Granzyme B markers. Median of each marker expression was calculated for each case by using quantitation from all Fields of View (FOV) that contained at least 10% tumor area. Results: AA tumors contained a significantly higher CD45-positive cells in overall, within tumor and stromal compartments (p = 0.0102, 0.0007 and 0.0280 respectively). Being more specific about CD45 marker, AA also showed to have a significantly higher level of overall lymphocyte population, measured by quantitating CD45+ CD14- cells (p =0.0081). AA tumors also had a significantly higher level of CD8 expression (p Conclusion: Significant differences are seen in the TME of AA vs. white TNBCs. Further characterization of macrophages, fibroblasts, regulatory T-cells and Extracellular Matrix are underway to more specifically define TME and determine the relationship between TME features and outcome. Citation Format: Vesal Yaghoobi, Vasiliki Pelekanou, Tess O'Meara, Andrea Silber, Lajos Pusztai, David Rimm, Kim Blenman. Comparison of tumor immune microenvironment in triple negative breast cancer (TNBC) of African American versus that of white patients [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4973.
- Published
- 2020
- Full Text
- View/download PDF
47. Phase II preoperative window study of SAR439859 versus letrozole in post-menopausal women with newly diagnosed estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer
- Author
-
Bethany Ling, Qiuyan Wang, Christina I. Herold, Sylvaine Cartot-Cotton, Vasiliki Pelekanou, and Mario Campone
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Standard of care ,business.industry ,Letrozole ,Endocrine therapy ,Estrogen receptor ,Newly diagnosed ,Post menopausal ,medicine.disease ,Breast cancer ,Internal medicine ,medicine ,business ,Human Epidermal Growth Factor Receptor 2 ,medicine.drug - Abstract
TPS1108 Background: Endocrine therapy targeting ER signaling is the standard of care for women with ER+ breast cancer. Selective ER degraders (SERDs) block ER signaling through dual competitive antagonism and receptor degradation. SAR439859, a potent, oral SERD, is in clinical development for ER+/HER2- breast cancer. This study is a 14-day preoperative non-therapeutic ‘window of opportunity’ trial to assess the direct effects of SAR439859 on tumor cell proliferation by evaluating the pharmacodynamic activity of SAR439859 in ER+/HER2- breast cancer. Methods: This international, open-label, Phase II randomized study (NCT04191382; ACT16106) evaluates SAR439859 at two dose levels versus the aromatase inhibitor letrozole by assigning 126 preoperative patients 1:1:1 to receive SAR439859 400 mg/day, SAR439859 200 mg/day or letrozole 2.5 mg/day. SAR439859 dosing is based on an ongoing Phase I/II study (NCT03284957; TED14856) in metastatic breast cancer (Campone. SABCS 2019. P5-11-02). Postmenopausal women with ER+/HER2- breast cancer indicated for immediate surgery (Stage I, Stage II or operable Stage III), Eastern Cooperative Oncology Group performance status 0–1 and Ki67 levels of ≥ 15% are eligible. Exclusion criteria include disorders potentially affecting absorption of SAR439859 or letrozole, and any prior therapy for breast cancer. Patients receive study treatment for 14 days, with the last dose given on the day before surgery. Paired tumor biopsies for assessment of biomarkers are performed at baseline and during surgery. The primary study endpoint is change in Ki67, a predictor of treatment benefit and long-term survival outcomes, after 14 days of treatment compared with baseline. Secondary endpoints include proportion of patients with ≥ 50% decrease in Ki67, ER expression to assess degradation, and safety. Pharmacokinetics of SAR439859, additional tumor markers, genomic mutation profile, Preoperative Endocrine Prognostic Index and pathological Complete Response will also be assessed. The study is currently recruiting; target enrollment: n = 126. Clinical trial information: NCT04191382 .
- Published
- 2020
- Full Text
- View/download PDF
48. Phase I/II study of SAR439859, an oral selective estrogen receptor degrader (SERD), in estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC)
- Author
-
Vasiliki Pelekanou, Hannah M. Linden, Alice Gosselin, Gary A. Ulaner, Sarat Chandarlapaty, Séverine Doroumian, Aditya Bardia, Mario Campone, and Marina Celanovic
- Subjects
Cancer Research ,business.industry ,Endocrine therapy ,Estrogen receptor ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,0302 clinical medicine ,Phase i ii ,Oncology ,030220 oncology & carcinogenesis ,Block (telecommunications) ,Cancer research ,Medicine ,business ,Human Epidermal Growth Factor Receptor 2 ,030215 immunology - Abstract
1070 Background: SERDs competitively antagonize and degrade the ER and can block signaling in ER-dependent tumors resistant to standard endocrine therapy (ET). This study (NCT03284957) investigates SAR439859, a potent oral SERD, in ER+/HER2- mBC. We present pooled dose escalation/expansion (Part A/B) data for SAR439859. Methods: Postmenopausal patients (pts) with ER+/HER2- mBC treated for ≥ 6 mos with prior ET received SAR439859 ≥ 150 mg QD (Part A) or 400 mg QD (Part B). Chemotherapy and targeted therapy in the advanced setting were allowed. Objective response rate (ORR; RECIST v1.1), clinical benefit rate (CBR; complete or partial response [PR] or stable disease [SD] ≥ 24 weeks), safety, and pharmacokinetics (PK) were assessed. Results: Pts (n = 62; Part A, 13; Part B, 49) had a median age of 63 yrs (range 37–88) and ECOG PS 0 (59.7%) or 1 (40.3%); 93.5% had visceral disease. All had prior ET, 74.2% had prior targeted therapy and 48.4% had ≥ 3 prior lines in the advanced setting. 61.3% of pts had treatment-related adverse events (TRAEs), all grade 1–2. Most frequent: hot flush (16.1%), constipation, arthralgia (both 9.7%), decreased appetite, vomiting, diarrhea, nausea (all 8.1%), fatigue (6.5%). No pts discontinued due to AEs. CBR was 35.6% overall, with antitumor activity irrespective of ESR1 mutation status (Table). In pts with no prior SERD, CDK4/6 or mTOR inhibitors (n = 14), ORR was 21.4% and CBR 64.3%. PK data for Part B and ESR1 mutation data will be provided. Conclusions: SAR439859 had a favorable safety profile with limited TRAEs. In these heavily pre-treated pts (prior targeted therapy in 74.2%), ORR and CBR were similar to historical fulvestrant performance in pts with no prior targeted therapy. Encouraging ORR and CBR in pts with no prior SERD, CDK4/6 or mTOR inhibitors (n = 14; ORR 21.4%; CBR 64.3%) supports SAR439859 development in earlier lines of therapy. Clinical trial information: NCT03284957 . [Table: see text]
- Published
- 2020
- Full Text
- View/download PDF
49. Phase II trial of SAR439859 vs endocrine monotherapy in pre- and post-menopausal, estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-), locally advanced or metastatic breast cancer (BC) with prior exposure to hormonal therapies
- Author
-
Vasiliki Pelekanou, Katharine Cuff, Arlene Chan, Qianying Liu, Sara M. Tolaney, Suzette Delaloge, Irfan Cicin, Amele Amrate, Sylvaine Cartot-Cotton, Peter A. Kaufman, Diego Kaen, and Rafael Betancourt
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Locally advanced ,Estrogen receptor ,medicine.disease ,Metastatic breast cancer ,Internal medicine ,Medicine ,Endocrine system ,In patient ,business ,Human Epidermal Growth Factor Receptor 2 ,Pre and post ,Hormone - Abstract
TPS1107 Background: Endocrine therapy (ET) targeting ER signaling is the mainstay of care for ER+ metastatic BC. There remains an unmet need in patients (pts) whose tumors become resistant to currently available ET. Selective ER degraders (SERDs) were developed to overcome resistance to existing ER-directed therapies by both competitively antagonizing and degrading ERs, while exploiting continued dependence of the tumor on ER signaling. SAR439859 is a potent SERD with robust preclinical ER degrading activity. In a Phase I dose escalation trial, SAR439859 demonstrated a favorable safety profile with no dose-limiting toxicities across all doses (20–600 mg QD). ER occupancy generally exceeded > 87% with plasma concentrations > 100 ng/mL. Overall response rate was 6.3% and clinical benefit rate was 50% (Campone. SABCS 2019. P5-11-02). The recommended Phase II monotherapy dose was 400 mg QD. Methods: This international, prospective, open-label, randomized Phase II study (NCT04059484; ACT16105) assesses safety and efficacy of SAR439859 in pts with ER+ (> 1%)/HER2- metastatic or locally advanced BC progressing on ≥ 6 months of continuous ET (0–2 lines in the metastatic setting). Prior CDK inhibitors are allowed. Exclusion criteria include Eastern Cooperative Oncology Group performance status (ECOG PS) ≥ 2, life expectancy < 3 months, > 1 chemotherapy or targeted therapy in the metastatic setting, concomitant illness and factors potentially affecting SAR439859 absorption. Pts are randomized 1:1 to SAR439859 400 mg QD orally or physician’s choice of endocrine monotherapy (fulvestrant, tamoxifen, aromatase inhibitor). Pts receive 28-day cycles until unacceptable toxicity, progression, death, investigator decision or pt request. Stratification factors include visceral metastases, prior CDK4/6 inhibitors, and ECOG PS. Primary endpoint is progression-free survival (RECIST v1.1). Secondary endpoints include overall survival, response rate, duration of response, clinical benefit, pharmacokinetics, quality of life and safety. Target enrollment: n = 282; current enrollment: n = 9. Funding: Sanofi Clinical trial information: NCT04059484 .
- Published
- 2020
- Full Text
- View/download PDF
50. Abstract PD5-01: Prospective multi-institutional evaluation of pathologist assessment of PD-L1 assays in triple negative breast cancer
- Author
-
Mary Ann Sanders, Anja C. Roden, Gang Han, Andrew M. Bellizzi, Vesal Yaghoobi, Hannah Wen, Dylan V. Miller, Omar Hameed, Vasiliki Pelekanou, Kamaljeet Singh, M G Kuba, Veerle Bossuyt, Amy Ly, Oluwole Fadare, Krisztina Z. Hanley, Jane E. Brock, Mirna Podoll, Fahad Shabbir Ahmed, Shi Wei, Kimberly Cole, Emily Reisenbichler, Lajos Pusztai, David L. Rimm, and Beth T. Harrison
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,Concordance ,Cancer ,medicine.disease ,Food and drug administration ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,Atezolizumab ,030220 oncology & carcinogenesis ,PD-L1 ,biology.protein ,Medicine ,business ,Lung cancer ,Triple-negative breast cancer - Abstract
Background: The IMpassion130 study demonstrated prolonged overall survival when atezolizumab, a PD-L1 inhibitor, was added to nab-paclitaxel in PD-L1 positive patients with advanced triple negative breast cancer (TNBC). Approximately 40% of tumors were PD-L1 positive, utilizing a cutoff of ≥ 1% immune cell (IC) staining with the Ventana SP142 immunohistochemical (IHC) assay. The Food and Drug Administration (FDA) approved this assay as a companion test to determine patient eligibility for atezolizumab therapy. However, literature in lung cancer shows that pathologists have a high discordance in assessing PD-L1 on IC. The FDA companion test documentation for SP142 shows high inter-laboratory reproducibility with nearly 95% overall percent agreement (OPA) between 2 readers for 2 category scoring of IC (positive versus negative) in TNBC in a central laboratory. PD-L1 testing for breast cancer is now becoming widespread in pathology laboratories. The goal of this study was to assess concordance in PD-L1 scoring between 19 pathologists from 14 institutions examining the same 86 TNBC tissues stained with the SP142 and SP263 antibodies. We also present a new method to determine the minimum number for evaluators needed to estimate concordance between large numbers of readers as occurs in real life setting. Methods: Full tissue sections were stained exactly as prescribed on the label on the Ventana Benchmark with the SP142 and SP263 PD-L1 assays. Sixty-eight and 76 cases were evaluable respectively. Stained slides were scanned and digital files distributed to pathologists who independently scored cases as either negative (< 1% IC staining) or positive (≥ 1% IC staining) and estimated the % of IC staining for positive cases. We applied a method that we call Observers Needed to Evaluate Subjective Tests (ONEST). For any combination of pathologists, we can quantify the degree of overall agreement using the proportion of tissue samples upon which all selected pathologists agreed, which can lead to a plot of non-increasing trajectory of agreement as the number of pathologists increases. We computed and plotted the empirical estimate of the overall agreement with the 95% confidence interval (CI) from 1000 randomly selected permutations of the pathologists. Results: PD-L1 was interpreted as positive with SP263 in an average of 78% of cases compared to 58% with the SP142 assay (p Citation Format: Emily Suzanne Reisenbichler, Gang Han, Vasiliki Pelekanou, Vesal Yaghoobi, Fahad S Ahmed, Andrew Bellizzi, Veerle Bossuyt, Jane Brock, Kimberly Cole, Oluwole Fadare, Omar Hameed, Krisztina Hanley, Beth T Harrison, M G Kuba, Amy Ly, Dylan Miller, Mirna Podoll, Anja C Roden, Mary A Sanders, Kamaljeet Singh, Shi Wei, Hannah Wen, Lajos Pusztai, David L Rimm. Prospective multi-institutional evaluation of pathologist assessment of PD-L1 assays in 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 PD5-01.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.