148 results on '"Seth A. Wander"'
Search Results
2. Interplay between ESR1/PIK3CA codon variants, oncogenic pathway alterations and clinical phenotype in patients with metastatic breast cancer (MBC): comprehensive circulating tumor DNA (ctDNA) analysis
- Author
-
Lorenzo Gerratana, Andrew A. Davis, Marko Velimirovic, Katherine Clifton, Whitney L. Hensing, Ami N. Shah, Charles S. Dai, Carolina Reduzzi, Paolo D’Amico, Firas Wehbe, Arielle Medford, Seth A. Wander, William J. Gradishar, Amir Behdad, Fabio Puglisi, Cynthia X. Ma, Aditya Bardia, and Massimo Cristofanilli
- Subjects
Circulating biomarker ,Endocrine therapy ,Next generation sequencing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background although being central for the biology and druggability of hormone-receptor positive, HER2 negative metastatic breast cancer (MBC), ESR1 and PIK3CA mutations are simplistically dichotomized as mutated or wild type in current clinical practice. Methods The study analyzed a multi-institutional cohort comprising 703 patients with luminal-like MBC characterized for circulating tumor DNA through next generation sequencing (NGS). Pathway classification was defined based on previous work (i.e., RTK, RAS, RAF, MEK, NRF2, ER, WNT, MYC, P53, cell cycle, notch, PI3K). Single nucleotide variations (SNVs) were annotated for their oncogenicity through OncoKB. Only pathogenic variants were included in the models. Associations among clinical characteristics, pathway classification, and ESR1/PIK3CA codon variants were explored. Results The results showed a differential pattern of associations for ESR1 and PIK3CA codon variants in terms of co-occurring pathway alterations patterns of metastatic dissemination, and prognosis. ESR1 537 was associated with SNVs in the ER and RAF pathways, CNVs in the MYC pathway and bone metastases, while ESR1 538 with SNVs in the cell cycle pathway and liver metastases. PIK3CA 1047 and 542 were associated with CNVs in the PI3K pathway and with bone metastases. Conclusions The study demonstrated how ESR1 and PIK3CA codon variants, together with alterations in specific oncogenic pathways, can differentially impact the biology and clinical phenotype of luminal-like MBC. As novel endocrine therapy agents such as selective estrogen receptor degraders (SERDS) and PI3K inhibitors are being developed, these results highlight the pivotal role of ctDNA NGS to describe tumor evolution and optimize clinical decision making.
- Published
- 2023
- Full Text
- View/download PDF
3. Adverse kidney outcomes of CDK 4/6 inhibitors for metastatic breast cancer
- Author
-
Paul E. Hanna, Ian A. Strohbehn, Daiana Moreno, Destiny Harden, Rituvanthikaa Seethapathy, Rani Sawtell, Qiyu Wang, Tianqi Ouyang, Nurit Katz-Agranov, James Dinulos, Seth A. Wander, Shruti Gupta, and Meghan E. Sise
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Cyclin-dependent kinase (CDK) 4/6 inhibitors have significantly improved overall and progression free survival of patients with metastatic breast cancer, but their effect on short and long-term kidney function is unknown. We found that early, mild estimated glomerular filtration rate (eGFR) decline was common in patients treated with CDK 4/6 inhibitors; however, severe kidney injury is rare and long-term eGFR decline is uncommon.
- Published
- 2023
- Full Text
- View/download PDF
4. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor-positive breast cancer
- Author
-
Jamie O. Brett, Laura M. Spring, Aditya Bardia, and Seth A. Wander
- Subjects
Breast cancer ,Hormone receptor/estrogen receptor ,ESR1 mutation ,Resistance ,Combination ,SERD ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract In metastatic hormone receptor-positive breast cancer, ESR1 mutations are a common cause of acquired resistance to the backbone of therapy, estrogen deprivation by aromatase inhibition. How these mutations affect tumor sensitivity to established and novel therapies are active areas of research. These therapies include estrogen receptor-targeting agents, such as selective estrogen receptor modulators, covalent antagonists, and degraders (including tamoxifen, fulvestrant, and novel agents), and combination therapies, such as endocrine therapy plus CDK4/6, PI3K, or mTORC1 inhibition. In this review, we summarize existing knowledge surrounding the mechanisms of action of ESR1 mutations and roles in resistance to aromatase inhibition. We then analyze the recent literature on how ESR1 mutations affect outcomes in estrogen receptor-targeting and combination therapies. For estrogen receptor-targeting therapies such as tamoxifen and fulvestrant, ESR1 mutations cause relative resistance in vitro but do not clearly lead to resistance in patients, making novel agents in this category promising. Regarding combination therapies, ESR1 mutations nullify any aromatase inhibitor component of the combination. Thus, combinations using endocrine alternatives to aromatase inhibition, or combinations where the non-endocrine component is efficacious as monotherapy, are still effective against ESR1 mutations. These results emphasize the importance of investigating combinatorial resistance, challenging as these efforts are. We also discuss future directions and open questions, such as studying the differences among distinct ESR1 mutations, asking how to adjust clinical decisions based on molecular surveillance testing, and developing novel therapies that are effective against ESR1 mutations.
- Published
- 2021
- Full Text
- View/download PDF
5. Immunogenicity of SARS-CoV-2 vaccines in patients with breast cancer
- Author
-
Elyssa Denault, Erika Nakajima, Vivek Naranbhai, Jennifer A. Hutchinson, Lindsey Mortensen, Elizabeth Neihoff, Caroline Barabell, Amy Comander, Dejan Juric, Irene Kuter, Theresa Mulvey, Jeffrey Peppercorn, Aron S. Rosenstock, Jennifer Shin, Neelima Vidula, Seth A. Wander, Beverly Moy, Leif W. Ellisen, Steven J. Isakoff, A. John Iafrate, Justin F. Gainor, Aditya Bardia, and Laura M. Spring
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose: To explore the immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in patients with breast cancer based on type of anticancer treatment. Methods: Patients with breast cancer had anti-spike antibody concentrations measured ⩾14 days after receiving a full SARS-CoV-2 vaccination series. The primary endpoint was IgA/G/M anti-spike antibody concentration. Multiple regression analysis was used to analyze log 10 -transformed antibody titer concentrations. Results: Between 29 April and 20 July 2021, 233 patients with breast cancer were enrolled, of whom 212 were eligible for the current analysis. Patients who received mRNA-1273 (Moderna) had the highest antibody concentrations [geometric mean concentration (GMC) in log 10 : 3.0 U/mL], compared to patients who received BNT162b2 (Pfizer) (GMC: 2.6 U/mL) (multiple regression adjusted p = 0.013) and Ad26.COV2.S (Johnson & Johnson/Janssen) (GMC: 2.6 U/mL) ( p = 0.071). Patients receiving cytotoxic therapy had a significantly lower antibody titer GMC (2.5 U/mL) compared to patients on no therapy or endocrine therapy alone (3.0 U/mL) ( p = 0.005). Patients on targeted therapies (GMC: 2.7 U/mL) also had a numerically lower GMC compared to patients not receiving therapy/on endocrine therapy alone, although this result was not significant ( p = 0.364). Among patients who received an additional dose of vaccine ( n = 31), 28 demonstrated an increased antibody response that ranged from 0.2 to >4.4 U/ mL. Conclusion: Most patients with breast cancer generate detectable anti-spike antibodies following SARS-CoV-2 vaccination, though systemic treatments and vaccine type impact level of response. Further studies are needed to better understand the clinical implications of different antibody levels, the effectiveness of additional SARS-CoV-2 vaccine doses, and the risk of breakthrough infections among patients with breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
6. Next-generation selective estrogen receptor degraders and other novel endocrine therapies for management of metastatic hormone receptor-positive breast cancer: current and emerging role
- Author
-
Maxwell R. Lloyd, Seth A. Wander, Erika Hamilton, Pedram Razavi, and Aditya Bardia
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Endocrine therapy (ET) is a pivotal strategy to manage early- and advanced-stage estrogen receptor-positive (ER+) breast cancer. In patients with metastatic breast cancer (MBC), progression of disease inevitably occurs due to the presence of acquired or intrinsic resistance mechanisms. ET resistance can be driven by ligand-independent, ER-mediated signaling that promotes tumor proliferation in the absence of hormone, or ER-independent oncogenic signaling that circumvents endocrine regulated transcription pathways. Estrogen receptor 1 ( ESR1 ) mutations induce constitutive ER activity and upregulate ER-dependent gene transcription, provoking resistance to estrogen deprivation and aromatase inhibitor therapy. The role ESR1 mutations play in regulating response to other therapies, such as the selective estrogen receptor degrader (SERD) fulvestrant and the available CDK4/6 inhibitors, is less clear. Novel oral SERDs and other next-generation ETs are in clinical development for ER+ breast cancer as single agents and in combination with established targeted therapies. Recent results from the phase III EMERALD trial demonstrated improved outcomes with the oral SERD elacestrant compared to standard anti-estrogen therapies in ER+ MBC after prior progression on ET, and other agents have shown promise in both the laboratory and early-phase clinical trials. In this review, we will discuss the emerging data related to oral SERDs and other novel ET in managing ER+ breast cancer. As clinical data continue to mature on these next-generation ETs, important questions will emerge related to the optimal sequence of therapeutic options and the genomic and molecular landscape of resistance to these agents.
- Published
- 2022
- Full Text
- View/download PDF
7. MicroRNA-Mediated Suppression of the TGF-β Pathway Confers Transmissible and Reversible CDK4/6 Inhibitor Resistance
- Author
-
Liam Cornell, Seth A. Wander, Tanvi Visal, Nikhil Wagle, and Geoffrey I. Shapiro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: CDK4/6 inhibition is now part of the standard armamentarium for patients with estrogen receptor-positive (ER+) breast cancer, so that defining mechanisms of resistance is a pressing issue. Here, we identify increased CDK6 expression as a key determinant of acquired resistance after palbociclib treatment in ER+ breast cancer cells. CDK6 expression is critical for cellular survival during palbociclib exposure. The increased CDK6 expression observed in resistant cells is dependent on TGF-β pathway suppression via miR-432-5p expression. Exosomal miR-432-5p expression mediates the transfer of the resistance phenotype between neighboring cell populations. Levels of miR-432-5p are higher in primary breast cancers demonstrating CDK4/6 resistance compared to those that are sensitive. These data are further confirmed in pre-treatment and post-progression biopsies from a parotid cancer patient who had responded to ribociclib, demonstrating the clinical relevance of this mechanism. Finally, the CDK4/6 inhibitor resistance phenotype is reversible in vitro and in vivo by a prolonged drug holiday. : Cornell et al. demonstrate a mechanism of acquired CDK4/6 inhibitor resistance that is independent of inherent genetic mutations, is conferred through extracellular signaling, and is reversible in vitro and in vivo. Resistance was mediated by exosomal miRNA, causing increased expression of CDK6 to overcome G1 arrest and promote cell survival. Keywords: breast cancer, drug resistance, targeted therapy, exosomes, CDK6, microRNA, TGF-β, SMAD4, palbociclib, ribociclib
- Published
- 2019
- Full Text
- View/download PDF
8. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia
- Author
-
Amir T. Fathi, Seth A. Wander, Traci M. Blonquist, Andrew M. Brunner, Philip C. Amrein, Jeffrey Supko, Nicole M. Hermance, Amity L. Manning, Hossein Sadrzadeh, Karen K. Ballen, Eyal C. Attar, Timothy A. Graubert, Gabriela Hobbs, Christelle Joseph, Ashley M. Perry, Meghan Burke, Regina Silver, Julia Foster, Meghan Bergeron, Aura Y. Ramos, Tina T. Som, Kaitlyn M. Fishman, Kristin L. McGregor, Christine Connolly, Donna S. Neuberg, and Yi-Bin Chen
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Aberrant expression of aurora kinase A is implicated in the genesis of various neoplasms, including acute myeloid leukemia. Alisertib, an aurora A kinase inhibitor, has demonstrated efficacy as monotherapy in trials of myeloid malignancy, and this efficacy appears enhanced in combination with conventional chemotherapies. In this phase I, dose-escalation study, newly diagnosed patients received conventional induction with cytarabine and idarubicin, after which alisertib was administered for 7 days. Dose escalation occurred via cohorts. Patients could then receive up to four cycles of consolidation, incorporating alisertib, and thereafter alisertib maintenance for up to 12 months. Twenty-two patients were enrolled. One dose limiting toxicity occurred at dose level 2 (prolonged thrombocytopenia), and the recommended phase 2 dose was established at 30mg twice daily. Common therapy-related toxicities included cytopenias and mucositis. Only three (14%) patients had persistent disease at mid-cycle, requiring “5+2” reinduction. The composite remission rate (complete remission and complete remission with incomplete neutrophil recovery) was 86% (nineteen of twenty-two patients; 90% CI 68–96%). Among those over age 65 and those with high-risk disease (secondary acute leukemia or cytogenetically high-risk disease), the composite remission rate was 88% and 100%, respectively. The median follow up was 13.5 months. Of those treated at the recommended phase 2 dose, the 12-month overall survival and progression-free survival were 62% (90% CI 33–81%) and 42% (90% CI 17–65%), respectively. Alisertib is well tolerated when combined with induction chemotherapy in acute myeloid leukemia, with a promising suggestion of efficacy. (clinicaltrials.gov Identifier:01779843).
- Published
- 2017
- Full Text
- View/download PDF
9. Triple negative breast cancer initiating cell subsets differ in functional and molecular characteristics and in γ‐secretase inhibitor drug responses
- Author
-
Diana J. Azzam, Dekuang Zhao, Jun Sun, Andy J. Minn, Prathibha Ranganathan, Katherine Drews‐Elger, Xiaoqing Han, Manuel Picon‐Ruiz, Candace A. Gilbert, Seth A. Wander, Anthony J. Capobianco, Dorraya El‐Ashry, and Joyce M. Slingerland
- Subjects
breast cancer stem cells ,GSI ,metastasis ,Notch1 ,Sox2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Increasing evidence suggests that stem‐like cells mediate cancer therapy resistance and metastasis. Breast tumour‐initiating stem cells (T‐ISC) are known to be enriched in CD44+CD24neg/low cells. Here, we identify two T‐ISC subsets within this population in triple negative breast cancer (TNBC) lines and dissociated primary breast cancer cultures: CD44+CD24low+ subpopulation generates CD44+CD24neg progeny with reduced sphere formation and tumourigenicity. CD44+CD24low+ populations contain subsets of ALDH1+ and ESA+ cells, yield more frequent spheres and/or T‐ISC in limiting dilution assays, preferentially express metastatic gene signatures and show greater motility, invasion and, in the MDA‐MB‐231 model, metastatic potential. CD44+CD24low+ but not CD44+CD24neg express activated Notch1 intracellular domain (N1‐ICD) and Notch target genes. We show N1‐ICD transactivates SOX2 to increase sphere formation, ALDH1+ and CD44+CD24low+cells. Gamma secretase inhibitors (GSI) reduced sphere formation and xenograft growth from CD44+CD24low+ cells, but CD44+CD24neg were resistant. While GSI hold promise for targeting T‐ISC, stem cell heterogeneity as observed herein, could limit GSI efficacy. These data suggest a breast T‐ISC hierarchy in which distinct pathways drive developmentally related subpopulations with different anti‐cancer drug responsiveness.
- Published
- 2013
- Full Text
- View/download PDF
10. Abstract PD13-09: PD13-09 Clinical outcomes of patients with HR+ HER2- advanced breast cancer with early progression on CDK4/6 inhibitors
- Author
-
Katherine K. Clifton, Shana N. Thomas, Jingqin Luo, Jing Xi, Nusayba A. Bagegni, Foluso O. Ademuyiwa, Rama Suresh, Ashley Frith, Andrew A. Davis, Ron Bose, Katherine Weilbaecher, Whitney L. Hensing, Timothy Pluard, Massimo Cristofanilli, Hyo S. Han, Adam M. Brufsky, Kevin Kalinsky, Shom Goel, Seth A. Wander, Lindsay L. Peterson, and Cynthia Ma
- Subjects
Cancer Research ,Oncology - Abstract
Background: CDK4/6 inhibitors (CDK4/6i) paired with endocrine therapy (ET) are considered first-line (1L) therapy for patients (pts) with HR+ HER2- advanced breast cancer (aBC). A minority of pts will demonstrate primary resistance to CDK4/6i, as characterized by early progression. Thymidine kinase 1 (TK1) is a cell-cycle regulated enzyme downstream of CDK4/6 and involved in nucleotide metabolism during DNA synthesis. Prior studies have shown TK1 may serve as a biomarker of response to CDK4/6i, with early TK1 activity (TK1a) suppression after initiation of CDK 4/6i therapy associated with improved PFS. Lack of TK1a suppression may be associated with primary resistance to CDK4/6i. In this study, we aim to analyze response to subsequent lines of therapy and overall survival (OS) of pts with early progression on 1L CDK4/6i. Methods: Pts with HR+ HER2- aBC from a phase II trial of an alternative schedule of palbociclib (palbo alt dosing trial NCT 3007979) and from a retrospective palbociclib study were included in this analysis. Pts in the palbo alt dosing trial underwent baseline and C1D15 TK1a analysis after initiation on CDK4/6i. C1D15 TK1a suppression was defined at TK1a < 30 Du/L. Pts in the retrospective palbociclib study included pts receiving palbo as part of their standard of care 1L therapy for HR+ HER2- aBC at Washington University in Saint Louis from 2016 to 2021. Clinical information, including treatment start and stop dates on each of the next-line therapies, were collected from the electronic medical record. PFS was estimated by the treatment duration on a specified treatment regimen. Early progression on CDK4/6i was defined as PFS < 6 mo. Best response was defined as next line of therapy with the numerically longest PFS. OS was defined as time to death from the initiation of CDK4/6i. Results: Of the 54 pts enrolled on the palbo alt dosing trial, 51 pts were evaluable for clinical benefit and 46 pts were evaluable for TK1a suppression rate at C1D15. 7 pts (15.2%) were found without TK1a suppression at C1D15. This lack of TK1a suppression on palbo was associated with a significantly shorter PFS (median PFS=3.1 mo) compared to not reached in pts with TK1a suppression at C1D15. We conducted clinical analysis on N=26 pts who exhibited early progression on CDK4/6i which included 10 pts from the palbo alt dosing trial and 16 from the retrospective study. The average subsequent line of therapies in this cohort was 3, with the most common second line (2L) therapy being chemotherapy (N=17, 65.4%) and ET (N=8, 30.8%). The median PFS for pts receiving 2L chemotherapy and ET was 4.09 mo and 3.64 mo, respectively. 10 pts received both chemotherapy and ET with 7 (70.0%) achieving best response with chemotherapy compared to 3 pts (30.0%) who achieved best response with ET. The median OS for the cohort was 14.6 mo. Conclusions: Early progression on CDK4/6i is associated with a particularly poor prognosis. In our cohort, the median OS was far below the expected median OS for pts receiving 1L palbo as reported in the PALOMA-2 trial (14.6 mo vs 53.9 mo). Early progression on CDK4/6i is associated with more aggressive disease which may respond more favorably to chemotherapy, as demonstrated by best response to therapy. Further prospective studies are warranted to explore this treatment approach. Citation Format: Katherine K. Clifton, Shana N. Thomas, Jingqin Luo, Jing Xi, Nusayba A. Bagegni, Foluso O. Ademuyiwa, Rama Suresh, Ashley Frith, Andrew A. Davis, Ron Bose, Katherine Weilbaecher, Whitney L. Hensing, Timothy Pluard, Massimo Cristofanilli, Hyo S. Han, Adam M. Brufsky, Kevin Kalinsky, Shom Goel, Seth A. Wander, Lindsay L. Peterson, Cynthia Ma. PD13-09 Clinical outcomes of patients with HR+ HER2- advanced breast cancer with early progression on CDK4/6 inhibitors [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD13-09.
- Published
- 2023
- Full Text
- View/download PDF
11. Abstract PD13-07: PD13-07 Combination therapy with the AKT inhibitor, ipatasertib, endocrine therapy, and a CDK4/6 inhibitor for hormone receptor positive (HR+)/HER2 negative metastatic breast cancer (MBC): results from the phase I TAKTIC trial
- Author
-
Seth A. Wander, Jennifer C. Keenan, Andrzej Niemierko, Dejan Juric, Laura M. Spring, Jeffrey Supko, Neelima Vidula, Steven J. Isakoff, Lianne Ryan, Sarah Padden, Elizabeth Fisher, Amber Newton, Beverly Moy, Leif Ellisen, Douglas S. Micalizzi, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
Background: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in combination with endocrine therapy (ET) provide significant clinical benefit in patients with HR+/HER2- metastatic breast cancer (MBC) and have become a standard of care treatment. Prior insights from tumor profiling and preclinical analyses suggest that AKT1 activation can induce CDK4/6i resistance. We hypothesized that targeting AKT1 following CDK4/6i progression may be an effective therapeutic strategy and conducted a clinical trial to evaluate both doublet (ET+AKTi) and triplet (ET+AKTIi+CDK 4/6i) therapy in the ≥ 2nd line MBC setting. Methods: TAKTIC is an open-label phase Ib clinical trial (clinicaltrials.gov NCT03959891) evaluating the combination of the AKT inhibitor ipatasertib (ipat) with fulvestrant (Arm A), an aromatase inhibitor (Arm B), or the triplet combination (Arm C) with fulvestrant + palbociclib (palbo). The primary objective is to evaluate the safety (NCI CTCAE 5.0) and tolerability of ipat in combination with endocrine therapy +/- CDK4/6i. Secondary objectives include clinical efficacy, as determined by objective response rate (RECIST v1.1), clinical benefit rate (CBR), progression-free survival (PFS), and overall survival (OS). Key inclusion criteria include unresectable HR+/HER2- MBC; at least 1 prior therapy for MBC including any CDK4/6i; up to 2 prior lines of chemotherapy for MBC (no limit on prior endocrine therapy). Here, we present an updated interim analysis from all study arms. Results: The trial completed accrual with 77 pts enrolled from June 2019 – February 2022, including 19 on Arm A, 16 on Arm B, and 42 on Arm C. Median age was 62 (range 32-88) and 65/77 pts (84%) received prior CDK4/6i (median no. of prior lines = 3, range 1-13). 56/77 pts (73%) had measurable disease at baseline and 50/77 pts (65%) had visceral metastases in the liver/lung (68% Arm A, 44% Arm B, 71% Arm C). Pts enrolled on Arms A and B received ipat at 400mg in combination with fulvestrant or an aromatase inhibitor, respectively. In Arm C, 27/42 pts enrolled into the dose escalation phase and received ipat + palbo at varying doses in combination with fulvestrant. Two DLTs were observed in the 300mg ipat + 125mg palbo cohort (grade 4 neutropenia ≥ 7 days). ET+400mg ipat + 100mg palbo was determined to be the recommended phase 2 dose (R2PD), and the remaining 15/42 pts on Arm C were treated at this dose level in the expansion phase. Treatment was well tolerated in all arms. Grade 3 and 4 toxicities included neutropenia (39/77, 50.6%), leukopenia (15/77, 19.5%), diarrhea (11/77, 14/3%), transaminitis (7/77, 9.1%), lymphopenia (6/77, 7.8%), rash (6/77, 7.8%), and thrombocytopenia (3/77, 3.9%). As of 6/28/2022, 16/77 pts remain on treatment. The median treatment duration for all pts is estimated at 6 months (range 0.5-39). Among the 56 pts with measurable disease, 11 had partial response (PR) and 32 had stable disease (SD) as the best response. CBR, defined as percentage of pts who achieved PR or SD > 6 months, was 48% across the study (53% Arm A, 31% Arm B, 57% Arm C). The median PFS was 5.5 months (95% confidence interval [CI]: 3.8 – 7.4) and the median OS was 24.5 months (95% CI: 17.1 – 33.9). Conclusions: The combination of ipat with endocrine therapy +/- palbo is well tolerated in heavily pre-treated pts, with preliminary evidence of clinical activity. This trial demonstrates how molecular insights related to CDK4/6i resistance inform potential therapy combinations. Further studies are needed to evaluate AKTi-based combinations in pts with HR+ MBC. Citation Format: Seth A. Wander, Jennifer C. Keenan, Andrzej Niemierko, Dejan Juric, Laura M. Spring, Jeffrey Supko, Neelima Vidula, Steven J. Isakoff, Lianne Ryan, Sarah Padden, Elizabeth Fisher, Amber Newton, Beverly Moy, Leif Ellisen, Douglas S. Micalizzi, Aditya Bardia. PD13-07 Combination therapy with the AKT inhibitor, ipatasertib, endocrine therapy, and a CDK4/6 inhibitor for hormone receptor positive (HR+)/HER2 negative metastatic breast cancer (MBC): results from the phase I TAKTIC trial [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD13-07.
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract PD1-10: Impact of Race on Clinical, Socioeconomic, and Genomic Characteristics, Clinical Trial Participation, and Receipt of Genotype-matched Therapy Among Patients with Metastatic Breast Cancer
- Author
-
Rupali Sood, Lianne Ryan, Andrzej Niemierko, Laura M. Spring, Dejan Juric, Steven J. Isakoff, Seth A. Wander, Jennifer Shin, Naomi Ko, Leif Ellisen, Beverly Moy, Aditya Bardia, and Neelima Vidula
- Subjects
Cancer Research ,Oncology - Abstract
Background: Clinical outcomes in breast cancer differ across racial and ethnic populations. We have previously demonstrated that receipt of genotype-matched therapy targeted to an actionable mutation may potentially improve patient outcomes (Vidula, CCR, 2021). We evaluated the impact of race on clinical, socioeconomic, and genomic characteristics, clinical trial participation, and receipt of genotype-matched therapy among patients with metastatic breast cancer (MBC). Methods: We conducted a retrospective study of patients with MBC at an academic institution who underwent cell-free DNA testing (cfDNA, Guardant360, 74 gene panel) as part of routine clinical care from 11/29/2016-11/2/2020. Patient demographics (including self-reported race and ethnicity) and clinical trial enrollment (at same institution) were determined by retrospective data collection. Mutations identified in cfDNA were characterized as actionable based on the variant interpretation performed by Guardant360 using vetted genomic databases, and receipt of genotype-matched therapy targeted to an actionable mutation was determined as previously described (Vidula, CCR, 2021). Pearson’s chi-squared and Wilcoxon rank-sum tests were used to compare categorical and continuous variables between groups, with p< 0.05 indicating statistical significance. Results: Four hundred and twenty-five patients with MBC and cfDNA results were identified, of which 369 were White (87%), 27 Black (6.4%), 15 Hispanic (3.5%), and 14 Asian (3.3%). There were no significant differences in median age at MBC diagnosis (p=0.064), disease subtype distribution (p=0.74), proportions of de-novo/recurrent MBC (p=0.95), presence of visceral metastases (p=0.84), Charleston comorbidity index (p=0.93), menopausal status (p=0.3), and level of education (p=0.44) across racial groups. Higher proportions of non-primary English speakers were seen in Hispanic (80%) and Asian (29%) races (p< 0.001). Median distance traveled to the institution also varied based on race, with White patients traveling further (White: 39.1 miles, Black: 21.8 miles, Hispanic 9.4 miles, Asian 9.1 miles, p< 0.001). In addition, type of insurance varied based on race, with White patients having the highest rates of commercial insurance and Medicare, Black patients having the highest rate of state-supported insurance, and Asian patients having the highest uninsured rates (p< 0.001). Clinical trial enrollment rates did not significantly differ by race (White: 44%, Black: 37%, Hispanic: 47%, and Asian 21%, p=0.34), but patients without insurance were significantly less likely to be enrolled on a trial than those with commercial insurance (p=0.03). The proportion of patients with ≥1 actionable mutation in cfDNA did not vary significantly by race (White: 78%, Black: 56%, Hispanic: 73%, Asian 86%, p=0.18) and the median number of actionable mutations found in cfDNA was similar across races (p=0.31). However, receipt of genotype-matched therapy targeted to an actionable mutation varied by race, with the highest rates of matched therapy in White patients (White: 28%, Black: 11%, Hispanic 13%, Asian 14%, p< 0.001). After multivariable logistic regression adjusting for subtype, commercial insurance versus other insurance types, and proximity to the center, White patients remained significantly more likely to receive matched therapy (p=0.029). Conclusions: We observed significant race-based differences in non-English speaking status, insurance type, and median distance traveled to the institution. Racial/ethnic minority patients were less likely to receive genotype-matched therapy than White patients. Further research is needed to identify barriers and reduce disparities in access to precision medicine. Citation Format: Rupali Sood, Lianne Ryan, Andrzej Niemierko, Laura M. Spring, Dejan Juric, Steven J. Isakoff, Seth A. Wander, Jennifer Shin, Naomi Ko, Leif Ellisen, Beverly Moy, Aditya Bardia, Neelima Vidula. Impact of Race on Clinical, Socioeconomic, and Genomic Characteristics, Clinical Trial Participation, and Receipt of Genotype-matched Therapy Among Patients with Metastatic Breast Cancer [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD1-10.
- Published
- 2023
- Full Text
- View/download PDF
13. Abstract P1-13-07: Investigating NF1 Mutations in Circulating Tumor DNA of Patients with Hormone-receptor Positive (HR+) Breast Tumors Resistant to CDK4/6 Inhibition (CDK4/6i): A Retrospective Clinical Analysis
- Author
-
Maxwell R. Lloyd, Lianne Ryan, Arielle J. Medford, Jennifer C. Keenan, Laura M. Spring, Neelima Vidula, Beverly Moy, Dejan Juric, Leif Ellisen, Aditya Bardia, and Seth A. Wander
- Subjects
Cancer Research ,Oncology - Abstract
Background: CDK4/6 inhibitors (CDK4/6i) are standard of care for the management of HR+/HER2- metastatic breast cancer (MBC). Genomic alterations that drive resistance to CDK4/6i are diverse, and while the molecular landscape is heterogeneous, several mechanisms of CDK4/6i resistance converge on the RAS/MAPK and PI3K/AKT/mTOR signaling pathways. NF1 downregulates RAS and dampens cellular proliferation. Laboratory-based models demonstrate that loss of NF1 is associated with resistance to endocrine therapy (ET), and emergence of NF1 mutations (NF1m) are correlated with progressive disease (PD) in circulating tumor DNA (ctDNA). While NF1m may diminish CDK4/6i susceptibility, a clear relationship has not been elucidated. The primary objective of this study was to characterize patient (pt) response to CDK4/6i in NF1m HR+/HER2- MBC. Methods: We identified 47 pts with NF1m via a database with one or more ctDNA samples sequenced at variable time-points as part of routine care for MBC. NF1m were categorized as pathogenic (p)NF1m or variants of uncertain significance (VUS) based on their associated Guardant report. We identified 27 pts with HR+/HER2- MBC and NF1m that received at least 1 line of CDK4/6i in the metastatic setting. Intrinsic resistance was defined as PD < 6 months on a CDK4/6i regimen, and acquired resistance was defined as PD >6 months. Pts with intrinsic resistance or acquired resistance and NF1m detected post-PD were categorized as having a resistance phenotype potentially driven by NF1m. Pts with NF1m detected prior to therapy and >6 months clinical response on a CDK4/6i were categorized as having NF1m tumors sensitive to CDK4/6i. Results: The NF1m cohort (n = 27) had 9 pts with pNF1m, while 18 pts expressed VUS. The median age at MBC diagnosis was 54 years, and 67% had visceral metastasis at ctDNA collection. Pts received a median of 1 prior line (range: 0 - 6) of ET or chemotherapy in the metastatic setting before CDK4/6i. Amongst pts with pathogenic variants (n = 9), we found 3 pts with pNF1m were intrinsically resistant to CDK4/6i. Acquired resistance was seen in 1 pt with pNF1m detected post-PD, and 2 pts had evidence of both acquired and subsequent intrinsic resistance to a later line of CDK4/6i. Overall, 67% (6/9) of pNF1m pts demonstrated a CDK4/6i resistance phenotype; mutant allele fraction (AF) ranged from 0.2% - 29.9%, and the mean maximum allele fraction (MAF) was 6.0%. Pre- and post-treatment samples were available on 3 pts with pNF1m, and 1 of these pts had an AF rise from 2.7% to 12.3% when comparing ctDNA pre- and post-CDK4/6i. ctDNA from 4 of 6 resistant tumors harbored other putative drivers including alterations in FGFR, KRAS, PTEN, and RB. We identified 2 counter-examples of pNF1m tumors sensitive to CDK4/6i. These pts expressed relatively low NF1m AF, ranging 0.1% - 0.5% with a mean MAF 0.3%. Another pNF1m pt had intrinsic resistance to initial CDK4/6i but was sensitive to later-line CDK4/6i. In the subgroup of pts with VUS-NF1m (n = 18), a more mixed picture of resistance and sensitivity was seen. 8 pts had intrinsic or acquired resistance, 8 pts had NF1m tumors sensitive to CDK4/6i, and 1 pt had evidence of both; 61% (n = 11) of pts expressed alterations in other resistance mediating genes. 1 pt stopped therapy due to toxicity rather than PD. Conclusions: Our work demonstrates that tumor expression of pNF1m may be associated with CDK4/6i resistance in pts with HR+/HER2- MBC, and allele fraction could be predictive of drug susceptibility. Tumors harboring VUS had varied sensitivity, suggesting that some of these mutations may not be pathogenic, and counter-examples of pNF1m MBC benefiting from CDK4/6i plus ET highlight the complexities in predicting drug response based on single gene alteration. Future effort is warranted to explore the potential impact of NF1 on CDK4/6i resistance, as well as the potential role for therapies targeting the MAPK pathway in this patient population. Citation Format: Maxwell R. Lloyd, Lianne Ryan, Arielle J. Medford, Jennifer C. Keenan, Laura M. Spring, Neelima Vidula, Beverly Moy, Dejan Juric, Leif Ellisen, Aditya Bardia, Seth A. Wander. Investigating NF1 Mutations in Circulating Tumor DNA of Patients with Hormone-receptor Positive (HR+) Breast Tumors Resistant to CDK4/6 Inhibition (CDK4/6i): A Retrospective Clinical Analysis [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P1-13-07.
- Published
- 2023
- Full Text
- View/download PDF
14. Abstract PD13-05: PD13-05 Updated results of a Phase 1b study of gedatolisib plus palbociclib and endocrine therapy in women with hormone receptor positive advanced breast cancer: Subgroup analysis by PIK3CA mutation status
- Author
-
Robert Wesolowski, Hope Rugo, Erica Stringer-Reasor, Hyo S. Han, Jennifer M. Specht, E. Claire Dees, Peter Kabos, Ulka Vaishampayan, Seth A. Wander, Janice Lu, Keerthi Gogineni, Alexander I. Spira, Anne F. Schott, Maysa Abu-Khalaf, Pratima Nayak, Brian F. Sullivan, Igor Gorbatchevsky, and AND Rachel M. Layman
- Subjects
Cancer Research ,Oncology - Abstract
Background: Addition of PI3K/mTOR inhibitor after progression on CDK4/6 inhibitor (CDK4/6i) and endocrine therapy (ET) can potentially restore sensitivity to CDK4/6i and prevent adaptive activation of the PI3K/mTOR pathway. To evaluate this hypothesis, we conducted a Phase Ib study of gedatolisib (G), a dual inhibitor of PI3K/mTOR, palbociclib (P) a CDK4/6i, and ET (with letrozole [LET] or fulvestrant [FUL]) in women with hormone receptor positive (HR+)/HER2- advanced breast cancer (ABC). Manageable toxicity and preliminary antitumor activity were observed in 35 patients(pts) enrolled in the dose escalation portion of the study (Forero-Torres, ASCO 2018) and 103 pts enrolled in the expansion portion of the study (Layman, SABCS 2021). Here, we report updated efficacy and safety data and sub-group analysis by PIK3CA mutation status in the four expansion study arms with a March 3, 2022, data cut-off. Methods: Pts with HR+/HER2- ABC were treated in four expansion arms: A) G+P+LET as first-line treatment, B) G+P+FUL as 2nd line treatment in pts without prior CDK4/6i; C & D) G+P+FUL as 2nd or 3rd line therapy in pts with prior CDK4/6i. P, LET, and FUL were administered at standard doses. G 180 mg was intravenously administered weekly in Arms A, B, and C and three weeks on/one week off in Arm D. The primary endpoint was investigator assessed objective response rate (ORR). Secondary endpoints included safety, duration of response and progression free survival (PFS). Results: Of the 103 pts treated with G+P+ ET in the expansion arms (A-D), 100% had measurable disease at baseline, 71% (73/103) lacked PIK3CA mutations (wild type; WT), 27% (28/103) had PIK3CA-mutations (MT), 70% (72/103) had evidence of bone metastases, and 59% (61/103) had liver metastases. The most frequent grade 3 and 4 treatment related AEs (TRAE) with G+P+ET included neutropenia (63%), stomatitis (27%), rash (20%), fatigue (11%) and hyperglycemia (7%). Treatment discontinuation due to TRAEs was 6.5% in Arm A, 15.4% in Arm B, 9.4% in Arm C and 3.7% in Arm D. Efficacy data for each arm is presented in Table 1. Promising ORR and PFS were seen in all arms regardless of PIK3CA mutation status. In Arm D, ORR was 63% overall, 73% in PIK3CA-MT pts, and 60% in PIK3CA-WT pts. Median PFS in Arm D was 12.9 months with a median follow up of 29 months. 60% and 48% of pts in the PIK3CA-MT and PIK3CA-WT Arm D sub-groups, respectively, were progression free at 12 months. Conclusions: These preliminary data demonstrate promising activity of G+P+ET combination in pts who were CDK4/6i-naïve and in those whose disease progressed on or after CDK4/6i therapy regardless of PIK3CA mutation status. Encouraging results in CDK4/6i treatment naïve patients warrant further evaluation of gedatolisib in combination with CDK4/6i treatment in the front-line setting. Arm D results provide a strong basis for the initiated Phase 3 study (VIKTORIA-1) in pts whose disease progressed on or after CDK4/6i therapy. Table 1. Efficacy Data by Expansion Arms Citation Format: Robert Wesolowski, Hope Rugo, Erica Stringer-Reasor, Hyo S. Han, Jennifer M. Specht, E. Claire Dees, Peter Kabos, Ulka Vaishampayan, Seth A. Wander, Janice Lu, Keerthi Gogineni, Alexander I. Spira, Anne F. Schott, Maysa Abu-Khalaf, Pratima Nayak, Brian F. Sullivan, Igor Gorbatchevsky, AND Rachel M. Layman. PD13-05 Updated results of a Phase 1b study of gedatolisib plus palbociclib and endocrine therapy in women with hormone receptor positive advanced breast cancer: Subgroup analysis by PIK3CA mutation status [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD13-05.
- Published
- 2023
- Full Text
- View/download PDF
15. Targeting CDK4 and 6 in Cancer Therapy: Emerging Preclinical Insights Related to Abemaciclib
- Author
-
Seth A Wander, Neil O’Brien, Lacey M Litchfield, Declan O’Dea, Claudia Morato Guimaraes, Dennis J Slamon, and Shom Goel
- Subjects
Cancer Research ,CDK4 and 6 ,Tumor Suppressor Proteins ,Oncology and Carcinogenesis ,Aminopyridines ,Cyclin-Dependent Kinase 4 ,Breast Neoplasms ,Estrogens ,Cyclin-Dependent Kinase 6 ,abemaciclib ,Estrogen ,Oncology ,5.1 Pharmaceuticals ,Breast Cancer ,preclinical ,Humans ,Benzimidazoles ,Female ,Oncology & Carcinogenesis ,Mitogens ,Development of treatments and therapeutic interventions ,Protein Kinase Inhibitors ,antitumor ,Cancer - Abstract
Pharmacologic inhibitors of cyclin-dependent kinases 4 and 6 (CDK4 and 6) are approved for the treatment of subsets of patients with hormone receptor positive (HR+) breast cancer (BC). In metastatic disease, strategies involving endocrine therapy combined with CDK4 and 6 inhibitors (CDK4 and 6i) improve clinical outcomes in HR+ BCs. CDK4 and 6i prevent retinoblastoma tumor suppressor protein phosphorylation, thereby blocking the transcription of E2F target genes, which in turn inhibits both mitogen and estrogen-mediated cell proliferation. In this review, we summarize preclinical data pertaining to the use of CDK4 and 6i in BC, with a particular focus on several of the unique chemical, pharmacologic, and mechanistic properties of abemaciclib. As research efforts elucidate the novel mechanisms underlying abemaciclib activity, potential new applications are being identified. For example, preclinical studies have demonstrated abemaciclib can exert antitumor activity against multiple tumor types and can cross the blood-brain barrier. Abemaciclib has also demonstrated distinct activity as a monotherapeutic in the treatment of BC. Accordingly, we also discuss how a greater understanding of mechanisms related to CDK4 and 6 blockade highlight abemaciclib’s unique in-class properties, and could pave new avenues for enhancing its therapeutic efficacy.
- Published
- 2022
- Full Text
- View/download PDF
16. Cyclin-Dependent Kinase 4/6 Inhibitors Beyond Progression in Metastatic Breast Cancer: A Retrospective Real-World Biomarker Analysis
- Author
-
Lorenzo Gerratana, Andrew A. Davis, Marko Velimirovic, Carolina Reduzzi, Katherine Clifton, Leslie Bucheit, Whitney L. Hensing, Ami N. Shah, Tania Pivetta, Charles S. Dai, Paolo D'Amico, Firas Wehbe, Arielle Medford, Seth A. Wander, William J. Gradishar, Amir Behdad, Cynthia X. Ma, Fabio Puglisi, Aditya Bardia, and Massimo Cristofanilli
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE As the continuation beyond progression (BP) of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is becoming increasingly attractive for the treatment of patients with hormone receptor (HR)–positive, human epidermal growth factor receptor 2 (HER2)–negative metastatic breast cancer (MBC), the definition of resistance factors is crucial. The aim of the study was to investigate the impact of CDK 4/6i BP and to explore potential genomic stratification factors. MATERIALS AND METHODS We retrospectively analyzed a multi-institutional cohort of patients with HR-positive HER2-negative MBC characterized for circulating tumor DNA through next-generation sequencing before treatment start. Differences across subgroups were analyzed by chi-square test, and survival was tested by univariable and multivariable Cox regression. Further correction was applied by propensity score matching. RESULTS Among the 214 patients previously exposed to CDK4/6i, 172 were treated with non–CDK4/6i-based treatment (non-CDK) and 42 with CDK4/6i BP. Multivariable analysis showed a significant impact of CDK4/6i BP, TP53 single-nucleotide variants, liver involvement, and treatment line on both progression-free survival (PFS) and overall survival (OS). Propensity score matching confirmed the prognostic role of CDK4/6i BP both for PFS and OS. The favorable impact of CDK4/6i BP was consistent across all subgroups, and a differential benefit was suggested for ESR1-mutated patients. ESR1 and RB1 mutations were more represented in the CDK4/6i BP subgroup with respect to CDK4/6i upfront. CONCLUSION The study highlighted a significant prognostic impact of the CDK4/6i BP strategy with a potential added benefit in patients with ESR1 mutations suggesting the need for an extensive biomarker characterization.
- Published
- 2023
- Full Text
- View/download PDF
17. A Gene Panel Associated With Abemaciclib Utility in ESR1-Mutated Breast Cancer After Prior Cyclin-Dependent Kinase 4/6-Inhibitor Progression
- Author
-
Jamie O. Brett, Taronish D. Dubash, Gabriela N. Johnson, Andrzej Niemierko, Veronica Mariotti, Leslie S.L. Kim, Jing Xi, Apurva Pandey, Siobhan Dunne, Azadeh Nasrazadani, Maxwell R. Lloyd, Avinash Kambadakone, Laura M. Spring, Douglas S. Micalizzi, Maristela L. Onozato, Dante Che, Utthara Nayar, Adam Brufsky, Kevin Kalinsky, Cynthia X. Ma, Joyce O'Shaughnessy, Hyo S. Han, Anthony J. Iafrate, Lianne Y. Ryan, Dejan Juric, Beverly Moy, Leif W. Ellisen, Shyamala Maheswaran, Nikhil Wagle, Daniel A. Haber, Aditya Bardia, and Seth A. Wander
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE For patients with hormone receptor–positive (HR+), human epidermal growth factor receptor 2–negative (HER2–) metastatic breast cancer (MBC), first-line treatment is endocrine therapy (ET) plus cyclin-dependent kinase 4/6 inhibition (CDK4/6i). After disease progression, which often comes with ESR1 resistance mutations (ESR1-MUT), which therapies to use next and for which patients are open questions. An active area of exploration is treatment with further CDK4/6i, particularly abemaciclib, which has distinct pharmacokinetic and pharmacodynamic properties compared with the other approved CDK4/6 inhibitors, palbociclib and ribociclib. We investigated a gene panel to prognosticate abemaciclib susceptibility in patients with ESR1-MUT MBC after palbociclib progression. METHODS We examined a multicenter retrospective cohort of patients with ESR1-MUT MBC who received abemaciclib after disease progression on ET plus palbociclib. We generated a panel of CDK4/6i resistance genes and compared abemaciclib progression-free survival (PFS) in patients without versus with mutations in this panel (CDKi-R[–] v CDKi-R[+]). We studied how ESR1-MUT and CDKi-R mutations affect abemaciclib sensitivity of immortalized breast cancer cells and patient-derived circulating tumor cell lines in culture. RESULTS In ESR1-MUT MBC with disease progression on ET plus palbociclib, the median PFS was 7.0 months for CDKi-R(–) (n = 17) versus 3.5 months for CDKi-R(+) (n = 11), with a hazard ratio of 2.8 ( P = .03). In vitro, CDKi-R alterations but not ESR1-MUT induced abemaciclib resistance in immortalized breast cancer cells and were associated with resistance in circulating tumor cells. CONCLUSION For ESR1-MUT MBC with resistance to ET and palbociclib, PFS on abemaciclib is longer for patients with CDKi-R(–) than CDKi-R(+). Although a small and retrospective data set, this is the first demonstration of a genomic panel associated with abemaciclib sensitivity in the postpalbociclib setting. Future directions include testing and improving this panel in additional data sets, to guide therapy selection for patients with HR+/HER2– MBC.
- Published
- 2023
- Full Text
- View/download PDF
18. Venous and arterial thrombosis associated with abemaciclib therapy for metastatic breast cancer
- Author
-
Nathan W. Watson, Seth A. Wander, Joseph J. Shatzel, and Hanny Al‐Samkari
- Subjects
Cancer Research ,Oncology ,Aminopyridines ,Anticoagulants ,Humans ,Benzimidazoles ,Breast Neoplasms ,Female ,Thrombosis ,Venous Thromboembolism - Abstract
The CDK4/6 inhibitor abemaciclib is a mainstay of treatment for hormone receptor-positive breast cancer. However, increased venous thromboembolism (VTE) rates in multiple clinical trials resulted in a black-box warning for this agent. Thrombosis rates in unselected real-world populations receiving abemaciclib remain ill defined.A multicenter observational cohort study was conducted of patients with metastatic breast cancer receiving abemaciclib. The primary end point was thrombosis during treatment or within 30 days of discontinuation. Multivariable logistic models assessed predictors of VTE, and a multivariable Cox proportional hazards model assessed mortality.A total of 364 patients were included, with a median treatment duration of 5.5 months. Twenty-six patients developed 27 (7.4%) thrombotic events (17 VTE, nine arterial thrombosis, and one with both events). No baseline characteristics were associated with increased VTE risk in multivariable modeling. Patients developing VTE during therapy had a higher risk of death than those who did not (hazard ratio, 2.09; 95% CI, 1.07-4.13). Median survival in patients who developed VTE compared with those who did not was 9.6 vs 25.8 months, respectively. The rate of VTE and any thrombosis during abemaciclib therapy was 9.1 and 13.7 events per 100 person-years, respectively, which is notably higher than rates observed in clinical trials.In a real-world setting, abemaciclib was associated with a VTE rate approximately two-fold greater than the already elevated rates reported in the MONARCH trials. Patients developing thrombosis on abemaciclib had a significantly higher risk of death. Given these findings, studies evaluating the role of thromboprophylaxis in patients receiving abemaciclib are needed.
- Published
- 2022
- Full Text
- View/download PDF
19. Abstract P1-14-02: Phase II study of adjuvant endocrine therapy with CDK 4/6 inhibitor, ribociclib, for localized ER+/HER2- breast cancer (LEADER, part 1)
- Author
-
Laura M Spring, Lauren Scarpetti, Andrzej Niemierko, Steven J Isakoff, Beverly Moy, Seth A Wander, Elisabeth Smith, Elizabeth Abraham, Jennifer Shin, Jaymin M Patel, Amy Comander, Therese Mulvey, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
Background: CDK 4/6 inhibitors have demonstrated substantial efficacy in treating ER+/HER2- metastatic breast cancer. Therefore, there is great interest in exploring their ability to reduce recurrence risk in early breast cancer. However, conflicting results were observed in the large adjuvant phase 3 clinical trials investigating combination of endocrine therapy and CDK 4/6 inhibitor (PALLAS, MONARCH-E). While these adjuvant clinical trials evaluated upfront use of CDK 4/6 inhibitor, the optimal timing of adding CDK 4/6 inhibitor for HR+/HER2- breast cancer remains unknown. We conducted a prospective phase II clinical trial to evaluate the addition of a CDK 4/6 inhibitor, ribociclib, in patients who were already on adjuvant endocrine therapy. Methods: In part 1 of the clinical trial, eligibility included patients with localized stage I-III ER+ (≥ 10%), HER2- breast cancer; completed surgery; and were on adjuvant endocrine therapy (any number of years) with at least one year or more of treatment remaining. Patients were randomized to two different ribociclib schedules: continuous (400 mg daily of 28-day cycle; arm 1) or intermittent (600 mg daily on days 1-21 of 28-day cycle; arm 2) for one year. Patients were concurrently treated with an aromatase inhibitor (plus GnRH agonist if premenopausal). Tolerance was evaluated via CTCAE version 4.03 and proportion of subjects who discontinued CDK 4/6 treatment early. Stratification factors for statistical analysis included: disease stage (III vs lower), duration of prior endocrine therapy (within 2 years; 2-5 years vs > 5 years), and whether the patient received prior chemotherapy or not. Baseline characteristics and risk factors for recurrence and for early discontinuation were compared between the arms of the study using Pearson's chi-squared test. Actuarial analysis of time to recurrence was done using the Kaplan-Meier estimator. The primary objective of part 1 was to estimate adherence to ribociclib treatment in the adjuvant setting. Results: In total, 81 patients were enrolled between February 2018 and September 2019, and 25 (31%) discontinued ribociclib treatment early, with no significant difference between study arms. The most common grade 3 or greater adverse events (AEs) leading to study discontinuation were neutropenia (44%), alanine aminotransferase increase (28%), and aspartate aminotransferase increase (16%). Among patients who discontinued early, neutropenia was more frequent in the 600 mg arm, 9 of 12 patients (75%), versus 2 of 13 patients (15%) in the 400 mg arm. No patients discontinued early due to prolonged QTc. Ribociclib was dose reduced for 22 patients (27%), with no significant difference between study arms (p = 0.12). After a median follow-up of 20 months, two patients have experienced disease recurrence with recurrence-free survival of 100% at 1 year and 97% (95% CI 88-99%) at 2 years. Biomarker (ctDNA) results will be reported at the meeting. Conclusions: Results demonstrate that while serious AEs with one year of adjuvant ribociclib are low, a substantial number of patients discontinued adjuvant CDK 4/6 inhibitor within 1 year. Overall, with limited follow-up, only two patients had recurrent disease since completion of ribociclib treatment. Tolerability and identifying patient subsets who will most benefit need to be carefully considered with CDK 4/6 inhibitors in the adjuvant setting. Citation Format: Laura M Spring, Lauren Scarpetti, Andrzej Niemierko, Steven J Isakoff, Beverly Moy, Seth A Wander, Elisabeth Smith, Elizabeth Abraham, Jennifer Shin, Jaymin M Patel, Amy Comander, Therese Mulvey, Aditya Bardia. Phase II study of adjuvant endocrine therapy with CDK 4/6 inhibitor, ribociclib, for localized ER+/HER2- breast cancer (LEADER, part 1) [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 P1-14-02.
- Published
- 2022
- Full Text
- View/download PDF
20. Abstract P3-23-02: Immunogenicity of SARS-CoV-2 vaccines in patients with breast cancer receiving CDK 4/6 inhibitors
- Author
-
Elyssa Denault, Erika Nakajima, Vivek Naranbhai, Alejandro Balazs, Lindsey Mortensen, Elizabeth Niehoff, Caroline Barabell, Jennifer A. Hutchinson, Seth A. Wander, Aron S. Rosenstock, Leif W. Ellisen, Beverly Moy, Steven J. Isakoff, Justin F. Gainor, A. John Iafrate, Aditya Bardia, and Laura M. Spring
- Subjects
Cancer Research ,Oncology - Abstract
Background: CDK 4/6 inhibitors have transformed the landscape of breast oncology. A CDK 4/6 inhibitor in combination with endocrine therapy is recommended as 1st line therapy for patients with metastatic hormone receptor positive breast cancer. CDK 4/6 inhibitors have purported immunomodulatory effects and while effective, myelosuppression is a common adverse effect of CDK 4/6 inhibitor treatment of breast cancer. The impact of CDK 4/6 inhibitor therapy on immunogenicity of vaccines is not known. In this study, we evaluated the spike antibody response to SARS-CoV-2 vaccines among patients with breast cancer receiving endocrine therapy with or without CDK 4/6 inhibitors.Methods: In the Cancer COVID and Vaccine (CANVAX) study eligible patients included patients with breast cancer who had completed all scheduled doses of SARS-CoV-2 vaccines. Chart review was conducted to identify patients who had received endocrine therapy with or without CDK 4/6 inhibitor. We used validated assays to measure anti-SARS-CoV-2 total IgA/M/G spike antibodies and virus neutralization. We evaluated the magnitude of antibody response based on geometric mean concentrations (GMCs) as well as the % of patients with inadequate seroconversion (defined as levels Citation Format: Elyssa Denault, Erika Nakajima, Vivek Naranbhai, Alejandro Balazs, Lindsey Mortensen, Elizabeth Niehoff, Caroline Barabell, Jennifer A. Hutchinson, Seth A. Wander, Aron S. Rosenstock, Leif W. Ellisen, Beverly Moy, Steven J. Isakoff, Justin F. Gainor, A. John Iafrate, Aditya Bardia, Laura M. Spring. Immunogenicity of SARS-CoV-2 vaccines in patients with breast cancer receiving CDK 4/6 inhibitors [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 P3-23-02.
- Published
- 2022
- Full Text
- View/download PDF
21. Abstract P2-07-02: Genomic predictors of rapid progression to first line endocrine and CDK4/6 inhibitor combination therapy in patients with estrogen receptor positive (ER+) HER-2 negative (HER2-) advanced breast cancer (ABC)
- Author
-
Marko Velimirovic, Lorenzo Gerratana, Andrew A Davis, Whitney L Hensing, Katherine Clifton, Ami N Shah, Paolo D'Amico, Charles S Dai, Elyssa N Denault, Cynthia X Ma, Seth A Wander, Dejan Juric, Massimo Cristofanilli, Bruce A Chabner, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
Background: Endocrine therapy with CDK 4/6 inhibitors (ET/CDK4/6i) represents the 1st line therapy for ER+/HER2- ABC. While majority of patients derive clinical benefit with combination therapy, a subset have refractory disease with progression within 6 months. However, predictive biomarkers for rapid progression are lacking. In this study, we evaluated genomic profiles associated with rapid disease progression on ET/CDK4/6i. Methods: We identified 77 patients who received 1st line ET/CDK4/6i combination therapy (AI or SERD with one of the 3 approved CDK4/6is) and had ctDNA analysis performed via plasma based genotyping utilizing the commercially available Guardant360 assay at three sites: Washington University in St. Louis, MO, Northwestern University (Chicago, IL), and Massachusetts General Hospital (Boston, MA). We aimed to look at the differences in patient characteristics and genomic profiles of the tumors assessed from baseline ctDNA specimens between the patients with rapid progression (time to progression TTP Citation Format: Marko Velimirovic, Lorenzo Gerratana, Andrew A Davis, Whitney L Hensing, Katherine Clifton, Ami N Shah, Paolo D'Amico, Charles S Dai, Elyssa N Denault, Cynthia X Ma, Seth A Wander, Dejan Juric, Massimo Cristofanilli, Bruce A Chabner, Aditya Bardia. Genomic predictors of rapid progression to first line endocrine and CDK4/6 inhibitor combination therapy in patients with estrogen receptor positive (ER+) HER-2 negative (HER2-) advanced breast cancer (ABC) [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 P2-07-02.
- Published
- 2022
- Full Text
- View/download PDF
22. Abstract P1-18-22: AKT inhibition in combination with endocrine therapy and a CDK4/6 inhibitor (CDK4/6i) in patients with hormone receptor positive (HR+)/HER2 negative metastatic breast cancer (MBC) and prior CDK4/6i exposure: A translational investigation
- Author
-
Seth A. Wander, Douglas S. Micalizzi, Taronish Dubash, Dejan Juric, Laura M. Spring, Neelima Vidula, Jennifer Keenan, Maureen Beeler, Elene Viscosi, Dante Che, Elizabeth L. Fisher, Rachel A. Hepp, Beverly Moy, Steven J. Isakoff, Leif W. Ellisen, Jeffrey G. Supko, Shyamala Maheswaran, Daniel A. Haber, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
Background: The cyclin-dependent kinase 4/6 inhibitors, with endocrine therapy (ET), have become the standard of care for patients with HR+/HER2- MBC. Prior insight from tumor biopsies and preclinical analyses suggest that AKT1 activation can provoke CDK4/6i resistance, highlighting a potential therapeutic role for AKT inhibition (AKTi) in this setting. However, combinatorial inhibition can be associated with significant toxicity and identification of the optimal biological dose is often challenging. In this translational co-clinical study, we evaluated escalating doses of AKTi combination with CDK 4/6i in parallel patient-derived pre-clinical models as well as a phase 1b clinical trial. Methods: In an open-label phase Ib dose-escalation clinical trial (TAKTIC, NCT03959891), we evaluated the safety, tolerability and efficacy of escalating doses of the AKT1 inhibitor ipatasertib (ipat) in combination with palbociclib (palbo) and fulvestrant (fulv) for patients with HR+/HER2- MBC. Inclusion criteria include unresectable or metastatic disease, at least 1 prior therapy for MBC including any CDK4/6i, and up to 2 prior lines of chemotherapy for MBC (no limit on prior endocrine therapy). In addition, response to escalating doses of ipat and palbo (with fulv) were explored in vitro via an ATP-based viability assay in tumor cell lines derived from circulating tumor cells (CTC) isolated from patients with endocrine-refractory HR+ MBC. Results: In the dose-escalation portion of the phase 1b clinical trial, 23 patients received the triplet combination of ipat, palbo, and fulv (median number of prior lines = 4.3, range 1-7; 100% with prior CDK4/6i): 3 pts received ipat at 200mg + 125mg palbo, 15 pts received 300mg + 125mg palbo, and 5 pts received 400mg + 100mg palbo, all with fulv (500 mg). Among the 23 patients, 20 patients (86.9%) had disease control (4 partial response and 16 stable disease) as the best response, per RECIST. Grade 3/4 toxicities included neutropenia (n=20), lymphopenia (n=3), diarrhea (n=3), thrombocytopenia (n=2), transaminitis (n=2), and rash (n=2). Two DLTs were observed in the 300mg ipat + 125mg palbo cohort (grade 4 neutropenia ≥ 7 days), but none at 400mg + 100mg palbo. The combination of ipat and palbo demonstrated an additive effect in vitro, with increased sensitivity to lower doses of palbo in the presence of ipat. Based on the totality of data, 400mg ipat + 100mg palbo + fulv 500 mg was selected as the recommended phase II dose (RP2D) in the post-CDK4/6i setting. Conclusions: The triplet combination of endocrine therapy with AKTi and lower dose CDK4/6i appears to be well tolerated in heavily pre-treated pts, with preliminary evidence of clinical activity. Further study is needed to evaluate biomarkers associated with higher AKTi benefit in order to guide rational development of combination therapy for patients with HR+/HER2- MBC in the post-CDK4/6i setting. Overall, this translational study demonstrates how insight into the molecular mechanisms of CDK4/6i resistance and combinatorial modeling can be leveraged to develop actionable therapeutic regimens for patients with MBC. Citation Format: Seth A. Wander, Douglas S. Micalizzi, Taronish Dubash, Dejan Juric, Laura M. Spring, Neelima Vidula, Jennifer Keenan, Maureen Beeler, Elene Viscosi, Dante Che, Elizabeth L. Fisher, Rachel A. Hepp, Beverly Moy, Steven J. Isakoff, Leif W. Ellisen, Jeffrey G. Supko, Shyamala Maheswaran, Daniel A. Haber, Aditya Bardia. AKT inhibition in combination with endocrine therapy and a CDK4/6 inhibitor (CDK4/6i) in patients with hormone receptor positive (HR+)/HER2 negative metastatic breast cancer (MBC) and prior CDK4/6i exposure: A translational investigation [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 P1-18-22.
- Published
- 2022
- Full Text
- View/download PDF
23. Supplemental Figure 2 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S2
- Published
- 2023
- Full Text
- View/download PDF
24. Supplemental Figure 9 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S9
- Published
- 2023
- Full Text
- View/download PDF
25. Data from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Mechanisms driving resistance to cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in hormone receptor–positive (HR+) breast cancer have not been clearly defined. Whole-exome sequencing of 59 tumors with CDK4/6i exposure revealed multiple candidate resistance mechanisms including RB1 loss, activating alterations in AKT1, RAS, AURKA, CCNE2, ERBB2, and FGFR2, and loss of estrogen receptor expression. In vitro experiments confirmed that these alterations conferred CDK4/6i resistance. Cancer cells cultured to resistance with CDK4/6i also acquired RB1, KRAS, AURKA, or CCNE2 alterations, which conferred sensitivity to AURKA, ERK, or CHEK1 inhibition. Three of these activating alterations—in AKT1, RAS, and AURKA—have not, to our knowledge, been previously demonstrated as mechanisms of resistance to CDK4/6i in breast cancer preclinically or in patient samples. Together, these eight mechanisms were present in 66% of resistant tumors profiled and may define therapeutic opportunities in patients.Significance:We identified eight distinct mechanisms of resistance to CDK4/6i present in 66% of resistant tumors profiled. Most of these have a therapeutic strategy to overcome or prevent resistance in these tumors. Taken together, these findings have critical implications related to the potential utility of precision-based approaches to overcome resistance in many patients with HR+ metastatic breast cancer.This article is highlighted in the In This Issue feature, p. 1079
- Published
- 2023
- Full Text
- View/download PDF
26. Supplemental Figure 8 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S8
- Published
- 2023
- Full Text
- View/download PDF
27. Supplemental Figure 5 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S5
- Published
- 2023
- Full Text
- View/download PDF
28. Supplemental Figure 4 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S4
- Published
- 2023
- Full Text
- View/download PDF
29. Supplemental Table 1 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 1
- Published
- 2023
- Full Text
- View/download PDF
30. Supplemental Table 2 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 2
- Published
- 2023
- Full Text
- View/download PDF
31. Supplemental Figure 7 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S7
- Published
- 2023
- Full Text
- View/download PDF
32. Supplemental Figure 10 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S10
- Published
- 2023
- Full Text
- View/download PDF
33. Supplemental Table 5 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 5
- Published
- 2023
- Full Text
- View/download PDF
34. Supplemental Table 7 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 7
- Published
- 2023
- Full Text
- View/download PDF
35. Supplemental Table 4 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 4
- Published
- 2023
- Full Text
- View/download PDF
36. Supplemental Figure 3 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S3
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Data from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplementary material
- Published
- 2023
- Full Text
- View/download PDF
38. Supplemental Table 8 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 8
- Published
- 2023
- Full Text
- View/download PDF
39. Supplemental Figure 6 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Figure S6
- Published
- 2023
- Full Text
- View/download PDF
40. Supplemental Table 3 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 3
- Published
- 2023
- Full Text
- View/download PDF
41. Supplemental Table 6 from The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor–Positive Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Sean G. Buchanan, Nancy U. Lin, Sara M. Tolaney, Eric P. Winer, Levi A. Garraway, Jill D. Kremer, Melissa E. Hughes, Gerald Batist, Quincy S. Chu, Gerard J. Oakley, Patricia S. Smith, John R. Stille, Valerie M. Jansen, Chunping Yu, Xiang S. Ye, Lacey M. Litchfield, Ricardo Martinez, Stephen H. Parsons, Adrienne G. Waks, Utthara Nayar, Kailey J. Kowalski, Karla Helvie, Pingping Mao, Flora Luo, Dewey Kim, Maxwell R. Lloyd, Jorge E. Buendia-Buendia, Gabriela N. Johnson, Xueqian Gong, Ofir Cohen, and Seth A. Wander
- Abstract
Supplemental Table 6
- Published
- 2023
- Full Text
- View/download PDF
42. Supplemental Table 11 from Acquired FGFR and FGF Alterations Confer Resistance to Estrogen Receptor (ER) Targeted Therapy in ER+ Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Nancy U. Lin, Eric P. Winer, Aviv Regev, David E. Root, Federica Piccioni, Vincent A. Miller, Orit Rozenblatt-Rosen, Samuel Freeman, Jon Chung, Utthara Nayar, Adrienne G. Waks, Seth A. Wander, Pedro Exman, Michael S. Cuoco, Jorge E. Buendia-Buendia, Justin G. Kusiel, Kailey J. Kowalski, Ofir Cohen, and Pingping Mao
- Abstract
Differentially expressed genes
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Table S2 from Tumor Tissue- versus Plasma-based Genotyping for Selection of Matched Therapy and Impact on Clinical Outcomes in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Leif W. Ellisen, Beverly Moy, Jerry Younger, Steven Isakoff, Dejan Juric, Laura Spring, Seth A. Wander, A. John Iafrate, Jochen Lennerz, Megan Yuen, Giuliana Malvarosa, Andrzej Niemierko, and Neelima Vidula
- Abstract
Supplemental Table S2. Characteristics of patients who underwent both cfDNA and tumor tissue genotyping testing.
- Published
- 2023
- Full Text
- View/download PDF
44. Supplementary Figure S1 from Tumor Tissue- versus Plasma-based Genotyping for Selection of Matched Therapy and Impact on Clinical Outcomes in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Leif W. Ellisen, Beverly Moy, Jerry Younger, Steven Isakoff, Dejan Juric, Laura Spring, Seth A. Wander, A. John Iafrate, Jochen Lennerz, Megan Yuen, Giuliana Malvarosa, Andrzej Niemierko, and Neelima Vidula
- Abstract
Supplemental Figure S1. Consort diagram of patients included in this study.
- Published
- 2023
- Full Text
- View/download PDF
45. Supplemental Table 9 from Acquired FGFR and FGF Alterations Confer Resistance to Estrogen Receptor (ER) Targeted Therapy in ER+ Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Nancy U. Lin, Eric P. Winer, Aviv Regev, David E. Root, Federica Piccioni, Vincent A. Miller, Orit Rozenblatt-Rosen, Samuel Freeman, Jon Chung, Utthara Nayar, Adrienne G. Waks, Seth A. Wander, Pedro Exman, Michael S. Cuoco, Jorge E. Buendia-Buendia, Justin G. Kusiel, Kailey J. Kowalski, Ofir Cohen, and Pingping Mao
- Abstract
RNA-seq experiment conditions and QC stats
- Published
- 2023
- Full Text
- View/download PDF
46. Supplemental Table 6 from Acquired FGFR and FGF Alterations Confer Resistance to Estrogen Receptor (ER) Targeted Therapy in ER+ Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Nancy U. Lin, Eric P. Winer, Aviv Regev, David E. Root, Federica Piccioni, Vincent A. Miller, Orit Rozenblatt-Rosen, Samuel Freeman, Jon Chung, Utthara Nayar, Adrienne G. Waks, Seth A. Wander, Pedro Exman, Michael S. Cuoco, Jorge E. Buendia-Buendia, Justin G. Kusiel, Kailey J. Kowalski, Ofir Cohen, and Pingping Mao
- Abstract
Evolutionary status data
- Published
- 2023
- Full Text
- View/download PDF
47. Supplemental Table 8 from Acquired FGFR and FGF Alterations Confer Resistance to Estrogen Receptor (ER) Targeted Therapy in ER+ Metastatic Breast Cancer
- Author
-
Nikhil Wagle, Nancy U. Lin, Eric P. Winer, Aviv Regev, David E. Root, Federica Piccioni, Vincent A. Miller, Orit Rozenblatt-Rosen, Samuel Freeman, Jon Chung, Utthara Nayar, Adrienne G. Waks, Seth A. Wander, Pedro Exman, Michael S. Cuoco, Jorge E. Buendia-Buendia, Justin G. Kusiel, Kailey J. Kowalski, Ofir Cohen, and Pingping Mao
- Abstract
Foundation Medicine cohort
- Published
- 2023
- Full Text
- View/download PDF
48. Data from Tumor Tissue- versus Plasma-based Genotyping for Selection of Matched Therapy and Impact on Clinical Outcomes in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Leif W. Ellisen, Beverly Moy, Jerry Younger, Steven Isakoff, Dejan Juric, Laura Spring, Seth A. Wander, A. John Iafrate, Jochen Lennerz, Megan Yuen, Giuliana Malvarosa, Andrzej Niemierko, and Neelima Vidula
- Abstract
Purpose:Actionable mutations can guide genotype-directed matched therapy. We evaluated the utility of tissue-based and plasma-based genotyping for the identification of actionable mutations and selection of matched therapy in patients with metastatic breast cancer (MBC).Experimental Design:Patients with MBC who underwent tissue genotyping (institutional platform, 91-gene assay) or plasma-based cell-free DNA (cfDNA, Guardant360, 73-gene assay) between January 2016 and December 2017 were included. A chart review of records to identify subtype, demographics, treatment, outcomes, and tissue genotyping or cfDNA results was performed. The incidence of actionable mutations and the selection of matched therapy in tissue genotyping or cfDNA cohorts was determined. The impact of matched therapy status on overall survival (OS) in tissue genotyping or cfDNA subgroups was determined with Cox regression analysis.Results:Of 252 patients who underwent cfDNA testing, 232 (92%) had detectable mutations, 196 (78%) had actionable mutations, and 86 (34%) received matched therapy. Of 118 patients who underwent tissue genotyping, 90 (76%) had detectable mutations, 59 (50%) had actionable mutations, and 13 (11%) received matched therapy. For cfDNA patients with actionable mutations, matched versus nonmatched therapy was associated with better OS [HR 0.41, 95% confidence interval (CI): 0.23–0.73, P = 0.002], and this remained significant in a multivariable analysis correcting for age, subtype, visceral metastases, and brain metastases (HR = 0.46, 95% CI: 0.26–0.83, P = 0.010).Conclusions:Plasma-based genotyping identified high rates of actionable mutations, which was associated with significant application of matched therapy and better OS in patients with MBC.See related commentary by Rugo and Huppert, p. 3275
- Published
- 2023
- Full Text
- View/download PDF
49. Data from FGFR1 Amplification Mediates Endocrine Resistance but Retains TORC Sensitivity in Metastatic Hormone Receptor–Positive (HR+) Breast Cancer
- Author
-
Aditya Bardia, Carlos L. Arteaga, A. John Iafrate, Leif W. Ellisen, Beverly Moy, Steven J. Isakoff, Dennis Sgroi, Giuliana Malvarosa, Megan Yuen, Jeffrey Peppercorn, Jerry Younger, Neelima Vidula, Laura M. Spring, Seth A. Wander, Alberto Servetto, Andrzej Niemierko, Dejan Juric, Luigi Formisano, and Joshua Z. Drago
- Abstract
Purpose:While FGFR1 amplification has been described in breast cancer, the optimal treatment approach for FGFR1-amplified (FGFR1+) metastatic breast cancer (MBC) remains undefined.Experimental Design: We evaluated clinical response to endocrine and targeted therapies in a cohort of patients with hormone receptor–positive (HR+)/HER2− MBC and validated the functional role of FGFR1-amplification in mediating response/resistance to hormone therapy in vitro.Results:In the clinical cohort (N = 110), we identified that patients with FGFR1+ tumors were more likely to have progesterone receptor (PR)-negative disease (47% vs. 20%; P = 0.005), coexisting TP53 mutations (41% vs. 21%; P = 0.05), and exhibited shorter time to progression with endocrine therapy alone and in combination with CDK4/6 inhibitor, but not with a mTOR inhibitor (everolimus), adjusting for key prognostic variables in multivariate analysis. Furthermore, mTOR-based therapy resulted in a sustained radiological and molecular response in an index case of FGFR1+ HR+/HER2− MBC. In preclinical models, estrogen receptor–positive (ER+)/FGFR1-amplified CAMA1 human breast cancer cells were only partially sensitive to fulvestrant, palbociclib, and alpelisib, but highly sensitive to everolimus. In addition, transduction of an FGFR1 expression vector into ER+ T47D cells induced resistance to fulvestrant that could be overcome by added TORC1 inhibition, but not PI3K or CDK4/6 inhibition.Conclusions:Collectively, these findings suggest that while FGFR1 amplification confers broad resistance to ER, PI3K, and CDK4/6 inhibitors, mTOR inhibitors might have a unique therapeutic role in the treatment of patients with ER+/FGFR1+ MBC.
- Published
- 2023
- Full Text
- View/download PDF
50. Supplementary Data from FGFR1 Amplification Mediates Endocrine Resistance but Retains TORC Sensitivity in Metastatic Hormone Receptor–Positive (HR+) Breast Cancer
- Author
-
Aditya Bardia, Carlos L. Arteaga, A. John Iafrate, Leif W. Ellisen, Beverly Moy, Steven J. Isakoff, Dennis Sgroi, Giuliana Malvarosa, Megan Yuen, Jeffrey Peppercorn, Jerry Younger, Neelima Vidula, Laura M. Spring, Seth A. Wander, Alberto Servetto, Andrzej Niemierko, Dejan Juric, Luigi Formisano, and Joshua Z. Drago
- Abstract
Supplementary figures
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.