277 results on '"Steven J, Isakoff"'
Search Results
2. Genomic spectrum of actionable alterations in serial cell free DNA (cfDNA) analysis of patients with metastatic breast cancer
- Author
-
Yael Bar, Jennifer C. Keenan, Andrzej Niemierko, Arielle J. Medford, Steven J. Isakoff, Leif W. Ellisen, Aditya Bardia, and Neelima Vidula
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract We aimed to study the incidence and genomic spectrum of actionable alterations (AA) detected in serial cfDNA collections from patients with metastatic breast cancer (MBC). Patients with MBC who underwent plasma-based cfDNA testing (Guardant360®) between 2015 and 2021 at an academic institution were included. For patients with serial draws, new pathogenic alterations in each draw were classified as actionable alterations (AA) if they met ESCAT I or II criteria of the ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT). A total of 344 patients with hormone receptor-positive (HR+)/HER2-negative (HER2-) MBC, 95 patients with triple-negative (TN) MBC and 42 patients with HER2-positive (HER2 + ) MBC had a baseline (BL) cfDNA draw. Of these, 139 HR+/HER2-, 33 TN and 13 HER2+ patients underwent subsequent cfDNA draws. In the HR+/HER2- cohort, the proportion of patients with new AA decreased from 63% at BL to 27–33% in the 2nd-4th draws (p
- Published
- 2024
- Full Text
- View/download PDF
3. Stressed target cancer cells drive nongenetic reprogramming of CAR T cells and solid tumor microenvironment
- Author
-
Yufeng Wang, David L. Drum, Ruochuan Sun, Yida Zhang, Feng Chen, Fengfei Sun, Emre Dal, Ling Yu, Jingyu Jia, Shahrzad Arya, Lin Jia, Song Fan, Steven J. Isakoff, Allison M. Kehlmann, Gianpietro Dotti, Fubao Liu, Hui Zheng, Cristina R. Ferrone, Alphonse G. Taghian, Albert B. DeLeo, Marco Ventin, Giulia Cattaneo, Yongxiang Li, Youssef Jounaidi, Peigen Huang, Cristina Maccalli, Hanyu Zhang, Cheng Wang, Jibing Yang, Genevieve M. Boland, Ruslan I. Sadreyev, LaiPing Wong, Soldano Ferrone, and Xinhui Wang
- Subjects
Science - Abstract
Abstract The poor efficacy of chimeric antigen receptor T-cell therapy (CAR T) for solid tumors is due to insufficient CAR T cell tumor infiltration, in vivo expansion, persistence, and effector function, as well as exhaustion, intrinsic target antigen heterogeneity or antigen loss of target cancer cells, and immunosuppressive tumor microenvironment (TME). Here we describe a broadly applicable nongenetic approach that simultaneously addresses the multiple challenges of CAR T as a therapy for solid tumors. The approach reprograms CAR T cells by exposing them to stressed target cancer cells which have been exposed to the cell stress inducer disulfiram (DSF) and copper (Cu)(DSF/Cu) plus ionizing irradiation (IR). The reprogrammed CAR T cells acquire early memory-like characteristics, potent cytotoxicity, enhanced in vivo expansion, persistence, and decreased exhaustion. Tumors stressed by DSF/Cu and IR also reprogram and reverse the immunosuppressive TME in humanized mice. The reprogrammed CAR T cells, derived from peripheral blood mononuclear cells of healthy donors or metastatic female breast cancer patients, induce robust, sustained memory and curative anti-solid tumor responses in multiple xenograft mouse models, establishing proof of concept for empowering CAR T by stressing tumor as a promising therapy for solid tumors.
- Published
- 2023
- Full Text
- View/download PDF
4. Treatment discontinuation, patient-reported toxicities and quality-of-life by age following trastuzumab emtansine or paclitaxel/trastuzumab (ATEMPT)
- Author
-
Tal Sella, Yue Zheng, Nabihah Tayob, Kathryn J. Ruddy, Rachel A. Freedman, Chau Dang, Denise Yardley, Steven J. Isakoff, Vicente Valero, Michelle DeMeo, Harold J. Burstein, Eric P. Winer, Antonio C. Wolff, Ian Krop, Ann H. Partridge, and Sara M. Tolaney
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract In the ATEMPT trial, adjuvant trastuzumab emtansine (T-DM1) compared to paclitaxel plus trastuzumab (TH) for stage I HER2-positive breast cancer improved patient-reported outcomes (PROs), while maintaining excellent disease outcomes. We report treatment discontinuation and use multivariable models to compare, patient-reported toxicity and quality-of-life (QOL) by age (≤50, >50) and treatment arm at 18 months post-enrollment among 366 eligible participants randomized in a 3:1 ratio to T-DM1 or TH. T-DM1 discontinuation was higher among women >50 vs. ≤50 (23% vs. 9%, p = 0.003, Fisher’s Exact test) with 4%, 8%, and 17% of older patients discontinuing treatment by 3, 6, and 9 months, respectively. Superior QOL with T-DM1 vs. TH was observed among women ≤50 with estimated mean difference of 6.48 (95% confidence interval (CI) 0.51–12.46) and driven by better social/family well-being and breast cancer-specific sub-scores. Among women >50, T-DM1 was associated with superior physical well-being and less activity impairment, with no differences in global QOL. Older women had decreased neuropathy with T-DM1 vs. TH. De-escalated treatment regimens for HER2 positive breast cancer may have age-varying impact on treatment tolerance, toxicities and subsequent QOL, which should be considered when selecting therapy options. Clinical Trial Registration: ClinicalTrials.gov, NCT01853748
- Published
- 2022
- Full Text
- View/download PDF
5. Cardiac outcomes of subjects on adjuvant trastuzumab emtansine vs paclitaxel in combination with trastuzumab for stage I HER2-positive breast cancer (ATEMPT) study (TBCRC033): a randomized controlled trial
- Author
-
Romualdo Barroso-Sousa, Paolo Tarantino, Nabihah Tayob, Chau Dang, Denise A. Yardley, Steven J. Isakoff, Vicente Valero, Meredith Faggen, Therese Mulvey, Ron Bose, Jiani Hu, Douglas Weckstein, Antonio C. Wolff, Katherine Reeder-Hayes, Hope S. Rugo, Bhuvaneswari Ramaswamy, Dan Zuckerman, Lowell Hart, Vijayakrishna K. Gadi, Michael Constantine, Kit Cheng, Frederick Briccetti, Bryan Schneider, Audrey Merrill Garrett, Kelly Marcom, Kathy Albain, Patricia DeFusco, Nadine Tung, Blair Ardman, Rita Nanda, Rachel C. Jankowitz, Mothaffar Rimawi, Vandana Abramson, Paula R. Pohlmann, Catherine Van Poznak, Andres Forero-Torres, Minetta Liu, Kathryn J. Ruddy, Yue Zheng, Shoshana M. Rosenberg, Richard D. Gelber, Lorenzo Trippa, William Barry, Michelle DeMeo, Harold Burstein, Ann Partridge, Eric P. Winer, Ian Krop, and Sara M. Tolaney
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The excellent outcomes seen in patients treated with adjuvant trastuzumab emtansine (T-DM1) in the ATEMPT trial and the favorable toxicity profile associated with this agent make T-DM1 a potential therapeutic option for select patients with stage I HER2-positive breast cancer. Moreover, T-DM1 is an established adjuvant treatment for patients with HER2-positive breast cancer with the residual invasive disease after neoadjuvant therapy. Given that cardiotoxicity is the most significant adverse event of trastuzumab, which is a main molecular component of T-DM1, we conducted a sub-analysis of the ATEMPT trial to determine the cardiac safety of adjuvant T-DM1. In this analysis, the incidence of grade 3–4 left ventricular systolic dysfunction (LVSD) in T-DM1 or trastuzumab plus paclitaxel arms were respectively 0.8 and 1.8%. In addition, three (0.8%) patients in the T-DM1 arm and six (5.3%) patients in the adjuvant paclitaxel with trastuzumab (TH) arm experienced a significant asymptomatic left ventricular ejection fraction (LVEF) decline that per-protocol required holding T-DM1 or trastuzumab. All patients with available follow-up data experienced full resolution of cardiac symptoms and LVEF normalization. Furthermore, we performed an exploratory analysis to assess the relationship between age, baseline LVEF, and body mass index with cardiac outcomes. No significant association between these baseline characteristics and the incidence of significant asymptomatic LVEF decline or symptomatic LVSD was identified. The low incidence of significant cardiac adverse events in this population during therapy with adjuvant T-DM1 suggests that studies on the cost-effectiveness of cardiac monitoring during adjuvant therapy using anthracycline-free regimens are needed. Clinical Trial Registration: ClinicalTrials.gov, NCT01853748
- Published
- 2022
- Full Text
- View/download PDF
6. Adjuvant trastuzumab and vinorelbine for early-stage HER2+ breast cancer
- Author
-
Shannon McLaughlin, Erika Nakajima, Yael Bar, Jennifer A. Hutchinson, Jennifer Shin, Beverly Moy, Steven J. Isakoff, Aditya Bardia, Irene Kuter, and Laura M. Spring
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: The single-arm phase II APT trial established trastuzumab and paclitaxel (TH) as the standard adjuvant regimen for small human epidermal growth factor receptor 2 (HER2+) tumors. However, paclitaxel causes alopecia and has high rates of neuropathy and hypersensitivity reactions. In patients with metastatic HER2+ breast cancer (BC), the combination of trastuzumab and vinorelbine (TV) is effective and well tolerated. There is a need for alternative non-anthracycline/taxane-based regimens for patients with HER2+ early-stage BC, especially for those with contraindications or who wish to avoid side effects of taxane-based regimens. Here we describe our institutional experience with adjuvant TV for patients with early-stage HER2+ BC. Methods: Clinicopathological characteristics, treatment details, and outcomes of patients with localized HER2+ BC treated with adjuvant TV from 2007 to 2021 at a large academic medical institution were collected. Study endpoints included invasive disease-free survival (IDFS), overall survival (OS), and safety/tolerability. IDFS and OS were measured from start date of TV treatment to date of event/last follow-up and date of death/last follow-up, respectively. Results: A total of 30 patients were treated with TV. All patients received trastuzumab at standard dosing and vinorelbine at a starting dose of 25 mg/m 2 either on days 1/8 or on days 1/8/21 (weekly) of a 21-day cycle with four planned cycles. Median age at diagnosis was 59 years (range: 36–81). 90.3% of patients had anatomic pathologic stage IA BC and 9.7% stage IIA BC. Of the 30 patients, 24 of them opted to pursue TV due to concerns related to alopecia, neuropathy, and other toxicities, and 6 switched from treatment with TH to TV due to toxicities. Eight patients experienced neutropenia with no cases of febrile neutropenia. No patients experienced alopecia or long-term neuropathy. With a median follow-up of 68 months (5.7 years), the 5-year IDFS rate was 90.9%, with one local and one distant recurrence. The 5-year OS was 100%. Conclusions: Trastuzumab in combination with vinorelbine in the adjuvant, early-stage setting for low-risk HER2+ BC demonstrated clinical efficacy and appeared to be well tolerated. TV warrants further evaluation as an alternative regimen to TH for patients with early-stage HER2+ BC.
- Published
- 2023
- Full Text
- View/download PDF
7. Phase 1b clinical trial of ado-trastuzumab emtansine and ribociclib for HER2-positive metastatic breast cancer
- Author
-
Laura M. Spring, Shealagh L. Clark, Tianyu Li, Shom Goel, Nabihah Tayob, Elene Viscosi, Elizabeth Abraham, Dejan Juric, Steven J. Isakoff, Erica Mayer, Beverly Moy, Jeffrey G. Supko, Sara M. Tolaney, and Aditya Bardia
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Patients with HER2+ metastatic breast cancer are often treated with a multitude of therapies in the metastatic setting, and additional strategies to prolong responses to anti-HER2 therapies are needed. Preclinical evidence suggests synergy between cyclin-dependent kinase 4 and 6 (CDK 4/6) inhibitors and anti-HER2 therapies. We conducted a phase 1b study of ribociclib and ado-trastuzumab emtansine (T-DM1) in patients with advanced/metastatic HER2-positive breast cancer previously treated with trastuzumab and a taxane in any setting, with four or fewer prior lines of therapy in the metastatic setting. A standard 3 + 3 dose-escalation design was used to evaluate various doses of ribociclib in combination with T-DM1, starting at 300 mg. The primary objective was to determine the maximum tolerated dose and/or recommended phase 2 dose (RP2D) of ribociclib in combination with T-DM1. A total of 12 patients were enrolled. During dose-escalation, patients received doses of ribociclib of 300 mg (n = 3), 400 mg (n = 3), 500 mg (n = 3), and 600 mg (n = 3). No dose-limiting toxicities were observed. The majority of toxicities were Grade 1 and 2, and the most common Grade 3 toxicities were neutropenia (33%), leukopenia (33%), and anemia (25%). After a median follow-up of 12.4 months, the median PFS was 10.4 months (95% confidence interval, 2.7–19.3). Based on the pharmacokinetic analysis, adverse events, and dose reductions, 400 mg was determined to be the RP2D for ribociclib given on days 8–21 of a 21-day cycle with T-DM1.
- Published
- 2021
- Full Text
- View/download PDF
8. 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
9. An Initial Evaluation of Human Plasma cMLC-1: A Potential Protein Biomarker for Trastuzumab-Induced Cardiotoxicity, Breast Cancer Screening and Progression
- Author
-
Ling Yu, Read Allen, Lin Jia, Ting Sun, Steven J. Isakoff, Marielle Scherrer-Crosbie, Allison M. Kehlmann, Hui Zheng, Amy Ly, Charlotte S. Walmsley, Katherine Hesler, Ava N. Varasteh, Christopher J. Pinto, Daniel E. McLoughlin, Wenjin Wu, and Xinhui Wang
- Subjects
cardiac myosin light chain 1(cMLC-1) ,biomarkers ,trastuzumab-induced cardiotoxicity ,breast cancer screening ,breast cancer progression ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
BackgroundTrastuzumab is a targeted therapy for human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, trastuzumab-induced cardiotoxicity (TIC) has been reported when trastuzumab is administered to patients as a single agent or combined with anthracycline. Currently no means for detecting the early onset of TIC such as a protein biomarker is available. In this regard and based on promising results from a preliminary animal study, the potential of cardiac myosin light chain 1(cMLC-1) as a biomarker to predict TIC, screen patients for breast cancer and monitor tumor progression in breast cancer patients was evaluated.MethodsArchived plasma samples collected before and after trastuzumab treatment at various fixed time points from 15 HER2+ patients with or without cardiotoxicity, recently collected plasma samples from 79 breast cancer patients (40 HER2+, 39 HER2-), and 46 healthy donors were analyzed for cMLC-1 levels using an enzyme-linked immunosorbent assay (ELISA).ResultsAn elevated plasma cMLC-1 level was found to be associated with TIC in 3 out of 7 (43%) trastuzumab-treated HER2+ breast cancer patients. However, this study provided an opportunity for us to study plasma cMCL-1 levels in breast cancer patients. It was demonstrated that elevated plasma cMCL-1 is associated with breast cancer. The cutoff cMLC-1 concentration is estimated to be 44.99 ng/mL with a sensitivity of 59.49% (95%CI: 48.47%-69.63%) and specificity of 71.74% (95%CI: 57.45% -82.68%). We also found a noticeable but not significantly more elevated plasma cMCL-1 level in HER2- than in HER2+ breast cancer patients with the given sample sizes. As a result, improved sensitivity of 79.49% (95%CI: 64.47%-89.22%) with the specificity of 63.04% (95%CI:48.60%-75.48%) were obtained for cMLC-1 to predict HER2- breast cancer with the cutoff at 37.17 ng/mL. Moreover, this study determined that cMLC-1 level was significantly higher in patients with metastatic breast cancer than in patients with non-metastatic breast cancer.ConclusionsWhile the analysis of cMLC-1 levels in the plasma of a limited number of trastuzumab-treated HER2+ breast cancer patients failed to fully support its identification as a blood protein biomarker for predicting TIC, additional analyses of plasma cMLC-1 levels did significantly establish its correlations with breast cancer and disease progression. Our findings shed light on and filled, to some extent, the gap of knowledge of the potential of cMLC-1 as a blood protein biomarker for screening breast cancer and monitoring disease progression of breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
10. Integration of Physician and Nursing Professional Efforts to Deliver Supportive Scalp Cooling Care to Oncology Patients at Risk for Alopecia
- Author
-
Lindsay L. Peterson, Maryam Lustberg, Sara M. Tolaney, Mikel Ross, Elahe Salehi, and Steven J. Isakoff
- Subjects
Adverse effects ,Alopecia ,Chemotherapy ,Cold caps ,Quality of life ,Scalp cooling ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Scalp cooling (SC) is an effective and generally well-tolerated method for prevention of chemotherapy-induced alopecia (CIA). Initially studied in early-stage breast cancer, these devices have expanded US Food and Drug Administration (FDA) clearance in a broad range of solid tumors including ovarian, colorectal, and prostate. Introducing SC to eligible patients, including those distraught by concerns of CIA, requires an integrated effort on the part of the physician, nursing, and care manager medical team. This article presents a pragmatic workflow for collaborative efforts from physicians and allied health professionals in the USA to deliver supportive SC to reduce CIA in patients undergoing treatment regimens known to impact hair follicles. It further highlights the efforts required to identify appropriate patients, educate, and set expectations of patients. The supervisory role of the physician during the procedure, the nursing role in monitoring and documentation, and the post-procedure decision-making by the physician are also addressed. Lastly, it suggests that integrated physician and nursing efforts necessary for scalp cooling are similar to other care used in oncology.
- Published
- 2020
- Full Text
- View/download PDF
11. Feasibility of introducing a smartphone navigation application into the care of breast cancer patients (The FIONA Study)
- Author
-
Steven J. Isakoff, Maya R. Said, Agnes H. Kwak, Eva Glieberman, Emily A. O’Rourke, Amanda Stroiney, Laura M. Spring, Beverly Moy, Aditya Bardia, Nora Horick, and Jeffrey M. Peppercorn
- Subjects
Cancer Research ,Oncology - Abstract
Purpose Patients with breast cancer (BC) face complex medical information and decisions. The Outcomes4Me mobile app provides evidence-based BC education, symptom management tracking and clinical trial matching. This study sought to evaluate the feasibility of introducing this app into routine BC care. Methods In this pilot study among BC patients undergoing therapy at an academic cancer center, patients were followed for 12 weeks with survey administration and electronic health record (EHR) abstraction at baseline and completion. Feasibility was defined as 40% of patients engaging with the app 3 or more times during the study. Additional endpoints included app usability (system usability scale), patient care experience, symptom evaluation, and clinical trial matching. Results The study enrolled 107 patients from 6/01/2020 to 3/31/2021. Utilization of the app was deemed feasible with 60% of patients engaging with the app at least 3 times. SUS score of 70 indicated above average usability. New diagnosis and higher education level was associated with greater app engagement, with usability similar across all age groups. 41% of patients found the app helped track symptoms. Cognitive and sexual symptoms were infrequently reported, but were more frequently captured in the app than in the EHR. After using the app, 33% of patients reported increased interest in clinical trial enrollment. Conclusion Introducing the Outcomes4Me patient navigation app into routine BC care is feasible and may improve the patient experience. These results support further evaluation of this mobile technology platform to improve BC education, symptom management, and decision making. Clinical trial registry Clinicaltrials.gov registration #: NCT04262518
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract OT2-09-01: TBCRC-055: A Phase II Study of NirAparib, Dostarlimab, and Radiotherapy in Metastatic, PD-L1 Negative or Immunotherapy-Refractory Triple-Negative Breast Cancer (NADiR) – NCT04837209
- Author
-
Elizabeth Scott, Steven J. Isakoff, Alphonse G. Taghian, Jean Wright, Cesar Augusto Santa-Maria, Payal Shah, Neil Taunk, Carey Anders, Rachel Blitzblau, Gaorav Gupta, and Alice Ho
- Subjects
Cancer Research ,Oncology - Abstract
Background: Metastatic triple negative breast cancer (mTNBC) is associated with high recurrence and mortality rates. Prior studies have shown an immune checkpoint inhibitor (ICI) + chemotherapy improves progression-free survival for patients with PD-L1 positive mTNBC. There remains a need for treatment options in patients who do not respond to ICI or are PD-L1 negative. Preclinical data suggests that a PARP inhibitor (PARPi) may promote innate immune signaling, and combination with an ICI has shown a positive response in patients with mTNBC. Radiotherapy (RT) is a potent immune stimulator and used for local control in the setting of metastatic breast cancer. This phase II study (NCT04837209) proposes combination of PARPi, ICI, and RT to combat ICI resistance and improve response rates in patients with mTNBC who are PD-L1 negative or who have progressed on prior ICI. Methods: 32 patients with mTNBC defined as ER< 1%, PR< 1%, HER-2-neu 0-1+ by IHC or non-FISH-amplified or patients with metastatic HR+/HER2- breast cancer are anticipated to participate. Eligibility criteria for mTNBC patients includes those who are PD-L1 negative or have progressed on prior ICI. Eligibility criteria for HR+/HER2- patients is specific to those who harbor a deleterious BRCA1 or BRCA2 mutation with or without high tumor mutational burden (TMB). All trial patients should have at least 1 lesion amenable to RT and at least 1 measurable lesion that will not be radiated. Study treatment consists of 3-week cycles, with 500mg dostarlimab given on day 1 of each cycle through cycle 5, then 1000mg given every 6 weeks. RT (24 Gy) is delivered in 3 consecutive fractions starting day 1 of cycle 1. Niraparib (200mg) is dosed orally daily. Tumor biopsies are taken within 28 days pre-treatment, and at C3D1-8. Blood samples are taken at baseline and every odd cycle for cfDNA and PBMC analysis. The primary endpoint is to assess overall response rate as measured by RECIST v1.1 of the combination of niraparib, dostarlimab, and RT. Secondary objectives include assessing safety and toxicity, overall survival, progression free survival, and quality of life. Results: To date, this study has accrued 4 subjects, including 3 with mTNBC, and 1 with HR+/HER2-/BRCA mutant + TMB high mBC. The study is currently open at MGH and Sibley, and the addition of UPenn, Johns Hopkins, and Duke are in progress. Funding for this study was provided by GSK. GSK was provided the opportunity to review a preliminary version of this abstract for factual accuracy, but the authors are solely responsible for final content and interpretation. People with specific interest in the trial should reach out to Elizabeth Scott, Clinical Research Coordinator, at ecscott@mgh.harvard.edu. Citation Format: Elizabeth Scott, Steven J. Isakoff, Alphonse G. Taghian, Jean Wright, Cesar Augusto Santa-Maria, Payal Shah, Neil Taunk, Carey Anders, Rachel Blitzblau, Gaorav Gupta, Alice Ho. TBCRC-055: A Phase II Study of NirAparib, Dostarlimab, and Radiotherapy in Metastatic, PD-L1 Negative or Immunotherapy-Refractory Triple-Negative Breast Cancer (NADiR) – NCT04837209 [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 OT2-09-01.
- Published
- 2023
- Full Text
- View/download PDF
13. 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
14. Abstract P5-14-09: Comparison of cell-free DNA genomics of breast cancer associated-chest wall disease vs. age & subtype matched controls with metastatic breast cancer not involving the chest wall
- Author
-
Neelima Vidula, Lianne Ryan, Andrzej Niemierko, Dejan Juric, Steven J. Isakoff, Francys Verdial, Marjan Azin, Laura A. Petrillo, Beverly Moy, Leif Ellisen, Shadmehr Demehri, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
Background: Breast cancer associated-chest wall disease (CWD) clinically behaves in an aggressive manner. However, little is known about the genomics of CWD. Cell-free DNA (cfDNA) can identify oncogenic mutations in metastatic breast cancer (MBC). We hypothesized that the cfDNA genomics of CWD may vary from MBC cases without CWD. We compared the cfDNA genomics of patients with CWD to that of age and subtype matched MBC controls without CWD. Methods: Patients with MBC at an academic institution who underwent cfDNA testing (Guardant360, next generation sequencing (NGS), 74 gene assay) from 2/2016-2/2021 were identified. Patients with documented CWD (chest wall recurrence with nodules, ulceration, and/or skin metastases on imaging and/or examination) at the time of cfDNA testing (coinciding with MBC diagnosis) were included in the CWD cohort. Age and subtype matched MBC controls without CWD (CON) during the same time period with cfDNA results at MBC diagnosis were identified. A retrospective review was conducted to determine clinical features and cfDNA genomics of CWD and CON. A two-sample test of proportions was used to compare CWD to CON, with p< 0.05 for statistical significance. Results: Thirty-one patients with CWD and 63 CON were identified. Both groups were well-matched in median age at MBC diagnosis (CWD 57 vs. CON 59 years, p=0.93) and subtype distribution (CWD: TNBC 35%, HR+/HER2- 58%, HER2+ 6%; CON: TNBC 29%, HR+/HER2- 65%, HER2+ 6%, p=0.78). Patients also had similar racial distribution in both cohorts (p=0.57). Ninety percent (28/31) of CWD vs. 90% (57/63) of CON had ≥1 mutation detectable in cfDNA (p=0.62). Median number of cfDNA mutations for CWD was 4.5 (range 0-16) vs. 4 (range 0-21) in CON (p=0.32). The most common cfDNA mutations in CWD were TP53 (58%), NOTCH1 (19%), PIK3CA (16%), BRCA1 (13%), NF1 (13%), FGFR2 (13%), ESR1 (13%), STK11 (10%), NTRK1 (10%), APC (10%), and KIT (10%). In comparison, the most common cfDNA mutations in CON were TP53 (54%), PIK3CA (35%), ESR1 (14%), GATA3 (13%), and ATM (11%). Table 1 depicts mutations that varied between CWD and CON which were statistically significant in ≥1 cohort analyzed. In HR+/HER2- MBC, NOTCH1, STK11, NTRK1, DDR2, and NF1 were significantly more common in CWD than CON. For TNBC, NOTCH1 was numerically more common in CWD vs. CON (p=0.06). Conclusions: The cfDNA genomic spectrum of CWD varies from MBC that does not infiltrate the chest wall. Mutations that are associated with metastasis (NOTCH1), inhibition of tumor suppression (STK11), tumor migration (DDR2), proliferation (NTRK1), and endocrine resistance (NF1) were significantly more common in HR+/HER2- CWD than matched controls. Further research is needed to validate these findings and determine the impact of matched targeted therapies for CWD. Table 1 N/A: mutation not observed Citation Format: Neelima Vidula, Lianne Ryan, Andrzej Niemierko, Dejan Juric, Steven J. Isakoff, Francys Verdial, Marjan Azin, Laura A. Petrillo, Beverly Moy, Leif Ellisen, Shadmehr Demehri, Aditya Bardia. Comparison of cell-free DNA genomics of breast cancer associated-chest wall disease vs. age & subtype matched controls with metastatic breast cancer not involving the chest wall [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 P5-14-09.
- Published
- 2023
- Full Text
- View/download PDF
15. 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
16. Neoadjuvant study of niraparib in patients with HER2-negative, BRCA-mutated, resectable breast cancer
- Author
-
Laura M. Spring, Hyo Han, Minetta C. Liu, Erika Hamilton, Hanna Irie, Cesar A. Santa-Maria, James Reeves, Peng Pan, Ming Shan, Yongqiang Tang, Julie R. Graham, Sebastien Hazard, Leif W. Ellisen, and Steven J. Isakoff
- Subjects
Cancer Research ,Oncology - Abstract
This single-arm pilot study (NCT03329937) evaluated neoadjuvant niraparib antitumor activity and safety in patients with localized HER2-negative, BRCA-mutated breast cancer. Twenty-one patients received niraparib 200 mg once daily in 28-day cycles. After 2 cycles, tumor response (≥30% reduction from baseline) by MRI was 90.5% and 40.0% (6 of 15) of patients who received only niraparib (2–6 cycles) had pathological complete response; no new safety signals were identified. High niraparib intratumoral concentration was observed.
- Published
- 2022
- Full Text
- View/download PDF
17. Breast Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology
- Author
-
William J. Gradishar, Meena S. Moran, Jame Abraham, Rebecca Aft, Doreen Agnese, Kimberly H. Allison, Bethany Anderson, Harold J. Burstein, Helen Chew, Chau Dang, Anthony D. Elias, Sharon H. Giordano, Matthew P. Goetz, Lori J. Goldstein, Sara A. Hurvitz, Steven J. Isakoff, Rachel C. Jankowitz, Sara H. Javid, Jairam Krishnamurthy, Marilyn Leitch, Janice Lyons, Joanne Mortimer, Sameer A. Patel, Lori J. Pierce, Laura H. Rosenberger, Hope S. Rugo, Amy Sitapati, Karen Lisa Smith, Mary Lou Smith, Hatem Soliman, Erica M. Stringer-Reasor, Melinda L. Telli, John H. Ward, Kari B. Wisinski, Jessica S. Young, Jennifer Burns, and Rashmi Kumar
- Subjects
Carcinoma, Intraductal, Noninfiltrating ,Oncology ,Humans ,Breast Neoplasms ,Female ,Medical Oncology - Abstract
The therapeutic options for patients with noninvasive or invasive breast cancer are complex and varied. These NCCN Clinical Practice Guidelines for Breast Cancer include recommendations for clinical management of patients with carcinoma in situ, invasive breast cancer, Paget disease, phyllodes tumor, inflammatory breast cancer, and management of breast cancer during pregnancy. The content featured in this issue focuses on the recommendations for overall management of ductal carcinoma in situ and the workup and locoregional management of early stage invasive breast cancer. For the full version of the NCCN Guidelines for Breast Cancer, visitNCCN.org.
- Published
- 2022
- Full Text
- View/download PDF
18. Local Therapy Outcomes and Toxicity From the ATEMPT Trial (TBCRC 033): A Phase II Randomized Trial of Adjuvant Trastuzumab Emtansine Versus Paclitaxel in Combination With Trastuzumab in Women With Stage I HER2-Positive Breast Cancer
- Author
-
Jennifer R. Bellon, Nabihah Tayob, David D. Yang, Jordan Tralins, Chau T. Dang, Steven J. Isakoff, Michelle DeMeo, Harold J. Burstein, Ann H. Partridge, Eric P. Winer, Ian E. Krop, and Sara M. Tolaney
- Subjects
Cancer Research ,Radiation ,Oncology ,Radiology, Nuclear Medicine and imaging - 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 PD5-11: Pilot study to assess prolonged nightly fasting in breast cancer survivors (LONGFAST)
- Author
-
Elizabeth K. O'Donnell, Yael N. Shapiro, Amy Comander, Steven J. Isakoff, Beverly Moy, Laura Spring, Seth Wander, Irene Kuter, Jennifer Shin, Jerry Younger, Michelle Specht, Chryssanthi Kourniotis, Carol Sullivan, Loren Winters, Nora Horick, and Jeffrey Peppercorn
- Subjects
Cancer Research ,Oncology - Abstract
Background: Prior, retrospective analysis of nightly fasting among women with breast cancer suggests that fasting less than 13 hours per night may be associated with higher risk of breast cancer recurrence. Small studies suggest that fasting duration can influence inflammation, obesity, sleep, and other potential mediators of breast cancer recurrence risk. Prolonged overnight fasting is a simple, nonpharmacological behavioral intervention strategy that may be doable for most patients. We designed this pilot study to prospectively evaluate the feasibility of prolonged overnight fasting among breast cancer survivors. Methods: We designed a single-arm, pilot study to evaluate the feasibility of fasting for 13 hours overnight for a 12-week period among women with a history of early stage breast cancer (I to III) who had completed initial cancer therapy at least 6 months prior. Baseline and end of study assessments included measurements of body mass index (BMI), quality of life (QOL) (Functional Assessment of Cancer Therapy - General (FACT-G)), mood (Hospital Anxiety and Depression Scale (HADS)), fatigue (Functional Assessment of Chronic Illness Therapy (FACIT) - Fatigue), levels of physical activity (Godin Leisure-Time Exercise Questionnaire), and blood biomarkers (expanded lipid profile, hemoglobin A1c, C-reactive protein, interleukin-6, tumor necrosis factor alpha, leptin, adiponectin). Patient-reported outcome (PRO) surveys were also administered at 6 weeks. Feasibility was defined as ≥ 60% of participants documenting fasting in the food diary for 13 hours on at least 70% of nights during the study period. Changes in study measures from baseline were evaluated using Wilcoxon signed-rank tests. Results: Between July 2020 and January 2021, we enrolled 40 women with a history of breast cancer. Participants had a median age of 59.9 (range 34.9-76.3) and median time since diagnosis was 4.5 years (range 0.8-20.7). At baseline, BMI was normal (18.5-24.9) in 40.0%, overweight (25-29.9) in 37.5%, and obese (≥30) in 22.5%. Forty-two and a half percent had Stage I cancer, 42.5% stage II, and 15.0% stage III. Sixty-five percent were on hormonal therapy. Ninety-five percent of participants fasted ≥ 13 hours for at least 70% of study days (95% CI 83%-99%). At 6 weeks, there was a statistically significant improvement in anxiety (p=.0007). No other significant changes were seen in PROs. At 12 weeks, there were statistically significant improvements in BMI (p=.0072), anxiety (p=.0141), depression (p=.0048), and fatigue (p=.0105). There was no association between change in BMI during the study and baseline BMI category, age, or endocrine therapy. There was no significant change in overall QOL, physical activity levels, or blood biomarkers at 12 weeks. Conclusions: Prolonged overnight fasting is feasible in the breast cancer population and may improve BMI, mood, and fatigue without a detrimental effect on overall QOL. The data from this study support the need for a larger, longer randomized study of prolonged overnight fasting in the breast cancer population to further evaluate the effects on body composition, mood, QOL, metabolic markers, and risk of recurrence. Table 1.Impact of Prolonged Overnight Fasting among Breast Cancer SurvivorsStudy AssessmentMedian at baselineMedian at 12 weeksMedian within-participant changep-valueBody Mass Index (kg/m2)26.4225.80-0.380.0072HADS - Depression1.001.00-1.000.0048HADS - Anxiety4.504.00-0.500.0141FACIT - Fatigue47.5049.821.000.0105FACT-G - Quality of Life95.2096.840.910.4933Physical Activity Level40.5039.000.000.3340Hemoglobin A1c (mg/dL)5.455.400.000.2758High-density lipoprotein (mg/dL)72.0073.00-2.000.4688Low-density lipoprotein (mg/dL)92.0099.001.000.5626Total Cholesterol (mg/dL)193.00192.003.000.6569C-reactive protein (mg/L)1.500.90-0.100.1043Interleukin-6 (pg/mL)2.001.90-0.300.1213Tumor Necrosis Factor α (pg/mL)0.740.74-0.050.2898Adiponectin (ug/mL)12.0012.000.000.0682Leptin (ng/mL)7.158.30-0.100.8418 Citation Format: Elizabeth K. O'Donnell, Yael N. Shapiro, Amy Comander, Steven J. Isakoff, Beverly Moy, Laura Spring, Seth Wander, Irene Kuter, Jennifer Shin, Jerry Younger, Michelle Specht, Chryssanthi Kourniotis, Carol Sullivan, Loren Winters, Nora Horick, Jeffrey Peppercorn. Pilot study to assess prolonged nightly fasting in breast cancer survivors (LONGFAST) [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 PD5-11.
- Published
- 2022
- Full Text
- View/download PDF
21. 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
22. Abstract P5-13-21: Relationship between circulating tumor DNA and response to neoadjuvant niraparib in HER2-negative, BRCA-mutated breast cancer
- Author
-
Ming Shan, Laura Spring, Minetta C Liu, Erika Hamilton, Hanna Irie, Cesar A Santa-Maria, Steven J Isakoff, James Reeves, Leif W Ellisen, Lee P Lim, Kavita Garg, Caterina Bertucci, Bin Feng, Hailei Zhang, Kaiming Sun, Julie R Graham, Erin Hofstatter, and Hyo Han
- Subjects
Cancer Research ,Oncology - Abstract
Background: Survival advantages with neoadjuvant chemotherapy (NACT) have been observed in patients (pts) with locally advanced breast cancer (BC) who achieve pathological complete response (pCR). Predicting risk of early recurrence in pts without pCR remains difficult. Circulating tumor DNA (ctDNA) may be a biomarker of neoadjuvant therapy response and recurrence. This study applied tumor genotyping of peripheral blood in pts with HER2− tumor BRCA-mutated (tBRCAm) BC treated with neoadjuvant niraparib. The relationship between ctDNA and tumor response was also characterized, with a focus on circulating TP53 mutant allelic frequency (MAF). Methods: Blood was collected from 21 pts enrolled in the study (NCT03329937) evaluating neoadjuvant niraparib in pts with localized HER2− tBRCAm BC. Pts received niraparib 200 mg once daily for two 28-day cycles. After 2 cycles, pts underwent surgery or received up to 6 cycles of niraparib and/or NACT prior to surgery. Blood samples were collected at screening, Cycle 1 Day 28 (C1D28), Cycle 2 Day 28 (C2D28), and pre-surgery. Ultrasound (USG) was done after each cycle and MRI after Cycle 2. Next-generation sequencing of cell-free plasma DNA was carried out using a proprietary target capture and analysis (Paweletz, Clin Cancer Res 2016). Results: ctDNA and TP53 mutations (TP53m) were detected at screening in 10/21 pts (47.6%). Across all 21 pts there was a correlation between TP53 MAF and baseline tumor stage (T) (mean [standard deviation], T1 0 [0], T2 0.05 [0.09], and T3 0.21 [0.12]; P=0.005). Linear regression modelling showed a positive correlation between tumor volume and baseline TP53 MAF (R2=0.206, P=0.0386). Baseline TP53 MAF also appeared to be associated with advanced disease stage (Table 1). Depletion of detectable TP53m from baseline was evident by C1D28 and persisted to pre-surgery (Table 2). This corresponded with decreases in tumor volume at C1D28 and further decreases at C2D28. Of 11 pts analyzed, 6 pts with >90% tumor volume decreases by MRI or USG at C2D28 had sustained TP53m depletion. Three pts had pCR; of these, 2 had sustained TP53m depletion (data unavailable for third pCR pt). Two pts with less robust tumor responses had increased pre-surgery TP53 MAF. Conclusions: We report the potential utility of ctDNA as a predictive biomarker for neoadjuvant niraparib response in pts with HER2− tBRCAm BC. Although sample size limited analysis of genes other than TP53, baseline ctDNA strongly correlated with tumor volume, and TP53 MAF was associated with tumor volume and trended towards association with disease stage. Longitudinal analysis of TP53 MAF suggested a correlation with niraparib response. Further study of ctDNA dynamics during NACT and correlation with pCR and long-term clinical outcomes is warranted. Funding: GlaxoSmithKline (GSK) study 213355; NCT03329937. Medical writing support was provided by Fishawack Indicia, part of Fishawack Health, funded by GSK. Table 1.Baseline TP53 MAF association with disease stage and subtypeTP53 MAFDisease Stage (I–III)Disease SubtypeStage IStage IIStage IIIHR+TNBC(n=8)(n=10)(n=3)(n=6)(n=15)Mean (SD)0.01 (0.02)0.07 (0.12)0.10 (0.05)0.02 (0.02)0.06 (0.10)HR, hormone receptor; MAF, mutant allelic frequency; SD, standard deviation; TNBC, triple-negative breast cancer. Table 2.TP53 MAF correlation with tumor volume decrease by ultrasound and MRIOutcomeScreeningCycle 1 Day 28 Cycle 2 Day 28 Pre-Surgeryn11997TP53 MAF, mean, (SD)0.09 (0.11)000.01 (0.02)n11112Percent change in tumor volume from baseline by MRI, mean (SD)-N/A*−72.6 (22.5)N/A*Percent change in tumor volume from baseline by ultrasound, mean (SD)-−60.8 (24.0)−80.6 (21.8)−95.9 (4.3)*MRI was only performed at Cycle 2 Day 28. MAF, mutant allelic frequency; MRI, magnetic resonance imaging; N/A, not applicable; SD, standard deviation. Citation Format: Ming Shan, Laura Spring, Minetta C Liu, Erika Hamilton, Hanna Irie, Cesar A Santa-Maria, Steven J Isakoff, James Reeves, Leif W Ellisen, Lee P Lim, Kavita Garg, Caterina Bertucci, Bin Feng, Hailei Zhang, Kaiming Sun, Julie R Graham, Erin Hofstatter, Hyo Han. Relationship between circulating tumor DNA and response to neoadjuvant niraparib in HER2-negative, BRCA-mutated 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 P5-13-21.
- Published
- 2022
- Full Text
- View/download PDF
23. Abstract P4-12-08: Accuracy of Patient Self-Reported Breast Cancer Disease Characteristics Compared to the Medical Record in a Trial of the Outcomes4MeDigital Health App
- Author
-
Steven J Isakoff, Eva Glieberman, Maya Said, Agnes H. Kwak, Emily A. O’Rourke, Amanda Stroiney, Laura Spring, Beverly Moy, Aditya Bardia, Nora Horick, and Jeffrey Peppercorn
- Subjects
Cancer Research ,Oncology - Abstract
Background: Patients’ understanding of their breast cancer (BC) diagnosis is important in improving treatment adherence, shared decision-making, and clinical trial matching. However, studies have reported discrepancies between electronic medical record (EMR) and patient reported information. Using data collected from a pilot study of the Outcomes4Me patient empowerment and clinical trial matching App, we analyzed concordance of patient reported disease characteristics compared to EMR data. Methods: Data was analyzed from a single institution pilot study (NCT04262518) evaluating the feasibility of introducing the Outcomes4Me app into routine BC care. Eligibility included BC patients with any subtype or stage of invasive cancer presenting with a new diagnosis or for follow-up on active therapy. We compared patient reported characteristics within a study specific survey and/or the Outcomes4Me app for stage (metastatic or not metastatic), recurrence history, hormone receptor status, HER2, and surgery history with the data recorded in the EMR. All statistics were descriptive. We conducted the same comparison between patient reported clinical characteristics among real world users of the Outcome4Me app and EMR records downloaded by that cohort of patients. Results: Between June 2020 and December 2020, 107 patients were enrolled. Baseline demographics: 90% White, 4% Black, 3% Asian; 37% with a college degree, and 43% with post college education; 66% hormone positive/HER2-, 20% HER2+, and 13% triple negative BC; 31% were stage 4. Concordance between the survey or App questionnaire and the EMR is shown in the Table. Comparing EMR and survey data, 62% of patients matched on both HER2 and HR status, and 94% of patients matched with the EMR on metastatic and recurrence status. When surgery and treatment information was included with these features, only 57% of patients matched across all these characteristics. Similar concordance was observed between the App questionnaire and EMR. Excluding the 21% of patients reporting “unsure” HER2 status improved the concordance to 85%. Overall concordance of recurrent or metastatic status was higher than for receptor status. Despite the discordance between EMR and patient-reported disease information, 97% of patients reported that they somewhat or strongly understood their cancer diagnosis. A similar pattern of concordance between the App questionnaire and EMR was observed among a real-world cohort of 636 patients using the Outcomes4Me App who provided medical record access. Conclusion: Self report of hormone receptor and HER2 status had limited concordance with the EMR, in contrast to a high degree of accuracy for self-report of metastatic disease. The limited accuracy of self-report suggests a need for improved patient education regarding their cancer characteristics and a need for caution when relying on self-report for clinical trials matching and targeted patient education. The use of a digital platform that integrates self-report with medical record access may help address these critical needs impacting patient empowerment and care. Concordance Between App Questionnaire, Study Survey and EMR (% of patients)Disease Characteristic Matching CriteriaApp v. EMR (n=85)Survey v. EMR (n=107)App Real World Cohort (n=636)HER2 Status73%66%79%HR Status80%85%80%Combined Receptor Status67%62%73%Metastatic Status94%(n=79)97%(n=98)94%Surgery History83%(n=47)95%96%(n=310)Recurrent Status98%(n=48)97%(n=98)98%(n=411)Recurrent/Metastatic Status93%(n=42)94%(n=98)98%(n=411)Receptor/Recurrent Metastatic Status and Surgery History74%(n=33)57%(n=93)70%(n=201) Citation Format: Steven J Isakoff, Eva Glieberman, Maya Said, Agnes H. Kwak, Emily A. O’Rourke, Amanda Stroiney, Laura Spring, Beverly Moy, Aditya Bardia, Nora Horick, Jeffrey Peppercorn. Accuracy of Patient Self-Reported Breast Cancer Disease Characteristics Compared to the Medical Record in a Trial of the Outcomes4MeDigital Health App [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 P4-12-08.
- Published
- 2022
- Full Text
- View/download PDF
24. 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
25. Abstract P2-14-17: A phase 1b study of PVX-410 vaccine in combination with pembrolizumab in metastatic triple negative breast cancer (mTNBC)
- Author
-
Steven J Isakoff, Nadine M. Tung, Jun Yin, Nabihah Tayob, Joanne Parker, Julie Rosenberg, Aditya Bardia, Laura Spring, Hannah Park, Maya Collins, William T. Barry, Mariano Severgnini, Doris Peterkin, and Sara M. Tolaney
- Subjects
Cancer Research ,Oncology - Abstract
Background: Immunotherapy with checkpoint inhibition is active in mTNBC. Both pembrolizumab and atezolizumab are FDA approved for programmed cell death ligand 1 positive (PDL1+) mTNBC. Vaccines may further induce host immune response and enhance therapeutic activity of checkpoint inhibitors. PVX-410 (PVX) (OncoPep, Inc.) is a novel, HLA-A2 restricted, tetra-peptide vaccine, with 3 of its 4 antigens (XBP1[2 splice variants] and CD138) commonly overexpressed in TNBC. We present results from a phase 1b study evaluating the immune response, safety and tolerability, and clinical activity of PVX and pembrolizumab (PEM) in mTNBC. Methods: Eligibility for this phase 1b multi-center, single-arm study included HLA-A2+, PD-L1 unselected female patients (pts) ≥18 years with metastatic or inoperable locally advanced TNBC, measurable disease, and any number of prior therapies, including prior checkpoint inhibitor therapy. Pts received 6 doses of 800µg PVX emulsified in Montanide ISA 720 VG by subcutaneous injection co-administered with intramuscular Hiltonol weekly for 6 weeks (wks) followed by booster vaccine doses at wks 10 and 28, with concurrent intravenous 200 mg PEM every 3 wks starting with the second PVX dose. Therapy was given until progressive disease, unacceptable toxicity or a maximum of 24 months. Blood samples were scheduled for immune response assessment at baseline and at weeks 2, 5, 10, 28, and 52 post-treatment initiation. The primary objective was PVX- specific immune response at week 10. Immune response was defined as a ≥2-fold change over baseline in the proportion of CD3+CD8+ T cells that expressed IFNγ and the proportion of CD3+CD8+ T cells positive for PVX tetramers following an in vitro stimulation of PBMC with PVX peptides using a flow cytometric assay. Secondary objectives were immune response at wk 28, safety and tolerability, and clinical endpoints (RR, CBR, DCR, DoR, PFS, and OS). Results: Between 3/2018 and 8/2020, 19 pts enrolled. Median age was 62 yrs (range 46-79), with median 2 (range 0-9) lines of prior therapy for metastatic disease. Median disease-free interval among 16 pts with prior early TNBC was 3.3 years. Among 19 enrolled patients, 16 were available for analysis at the time of abstract submission. Among the 16, 10 pts were evaluable at week 10 and 7(70%) demonstrated a PVX specific immune response. There were 6 patients who progressed before week 10, of whom 3 (50%) had a positive immune response at the EOT visit. Immune response persisted in all evaluable pts assessed at week 28 (n=4). Immune response data for all evaluable patients will be updated at the presentation. Among 19 patients evaluable for safety analysis, the most common adverse events (AEs) attributable to PVX (grade ≥2) included: fatigue (21%), arthralgia (11%) injection site reaction (5 %) pain (5%) lymphocyte count decreased (5%), maculopapular rash (5%) and skin infection (5%) . There were two grade 3 AEs attributed to PEM (AST elevation, hyponatremia) and one grade 4 AE (ALT elevation). There were no grade 5 AEs. The clinical benefit rate (CR+PR+SD for ≥16 weeks) was 31.6% with no confirmed partial or complete responses. Best overall response was SD in 9 (47%) patients. Analysis of additional clinical endpoints including PFS and OS is ongoing and will be presented at the meeting. Conclusions: PVX plus PEM is safe with manageable toxicity in pts with mTNBC. No new unexpected adverse events were identified. Immune response data show PVX induces antigen-specific T cell expansion as observed by increases in PVX tetramer and IFN positive T cells. Clinical disease control was observed with a CBR of 31.6%. Based on these promising immune response results in this pretreated population, a phase 2 study with PVX+PEM in combination with standard chemotherapy in treatment naïve, PD-L1+ mTNBC is underway (NCT04634747). Citation Format: Steven J Isakoff, Nadine M. Tung, Jun Yin, Nabihah Tayob, Joanne Parker, Julie Rosenberg, Aditya Bardia, Laura Spring, Hannah Park, Maya Collins, William T. Barry, Mariano Severgnini, Doris Peterkin, Sara M. Tolaney. A phase 1b study of PVX-410 vaccine in combination with pembrolizumab in metastatic triple negative breast cancer (mTNBC) [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 P2-14-17.
- Published
- 2022
- Full Text
- View/download PDF
26. Data from Parallel Genomic Alterations of Antigen and Payload Targets Mediate Polyclonal Acquired Clinical Resistance to Sacituzumab Govitecan in Triple-Negative Breast Cancer
- Author
-
Leif W. Ellisen, Aditya Bardia, Gad Getz, Dejan Juric, Laxmi Parida, Steven J. Isakoff, James R. Stone, Avinash Kambadakone, Charlotte S. Walmsley, Elyssa Denault, Chaya Levovitz, Filippo Utro, Kahn Rhrissorrakrai, Raquel A. Jacobs, Kara Slowik, Brian P. Danysh, Daniel McLoughlin, Elizabeth E. Martin, Nayana Thimmiah, Ignaty Leshchiner, Sheng Sun, and James T. Coates
- Abstract
Sacituzumab govitecan (SG), the first antibody–drug conjugate (ADC) approved for triple-negative breast cancer, incorporates the anti-TROP2 antibody hRS7 conjugated to a topoisomerase-1 (TOP1) inhibitor payload. We sought to identify mechanisms of SG resistance through RNA and whole-exome sequencing of pretreatment and postprogression specimens. One patient exhibiting de novo progression lacked TROP2 expression, in contrast to robust TROP2 expression and focal genomic amplification of TACSTD2/TROP2 observed in a patient with a deep, prolonged response to SG. Analysis of acquired genomic resistance in this case revealed one phylogenetic branch harboring a canonical TOP1E418K resistance mutation and subsequent frameshift TOP1 mutation, whereas a distinct branch exhibited a novel TACSTD2/TROP2T256R missense mutation. Reconstitution experiments demonstrated that TROP2T256R confers SG resistance via defective plasma membrane localization and reduced cell-surface binding by hRS7. These findings highlight parallel genomic alterations in both antibody and payload targets associated with resistance to SG.Significance:These findings underscore TROP2 as a response determinant and reveal acquired SG resistance mechanisms involving the direct antibody and drug payload targets in distinct metastatic subclones of an individual patient. This study highlights the specificity of SG and illustrates how such mechanisms will inform therapeutic strategies to overcome ADC resistance.This article is highlighted in the In This Issue feature, p. 2355
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figures S1-S4; Supplementary Tables S1-S4 from Parallel Genomic Alterations of Antigen and Payload Targets Mediate Polyclonal Acquired Clinical Resistance to Sacituzumab Govitecan in Triple-Negative Breast Cancer
- Author
-
Leif W. Ellisen, Aditya Bardia, Gad Getz, Dejan Juric, Laxmi Parida, Steven J. Isakoff, James R. Stone, Avinash Kambadakone, Charlotte S. Walmsley, Elyssa Denault, Chaya Levovitz, Filippo Utro, Kahn Rhrissorrakrai, Raquel A. Jacobs, Kara Slowik, Brian P. Danysh, Daniel McLoughlin, Elizabeth E. Martin, Nayana Thimmiah, Ignaty Leshchiner, Sheng Sun, and James T. Coates
- Abstract
Figure S1, radiographic assessment of key lesions of patient MGH-18; Figure S2, supplementary genomic analysis for patient MGH-18; Figure S3, supporting data for mechanism of TROP2 T256R; Figure S4, TROP2 immunohistochemistry for metastatic lesions of MGH-18; Table S1, detailed cohort characteristics; Table S2, patient treatment histories; Table S3, supplementary mutational data for MGH-18; Table S4, antibodies used.
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figure 1 from Phase II Trial of Bicalutamide in Patients with Androgen Receptor–Positive, Estrogen Receptor–Negative Metastatic Breast Cancer
- Author
-
Tiffany A. Traina, Clifford A. Hudis, Kimberly N. Feigin, Sujata Patil, Dilip D. Giri, Arooj Akhtar, Joseph M. Gonzalez, Lamia F. Momen, Mary Ellen Moynahan, Michael Danso, Ashley S. Doane, Vered Stearns, Andres Forero, Lisle Nabell, Hope Rugo, Kimberly Blackwell, Lisa A. Carey, Minetta C. Liu, James N. Ingle, Steven J. Isakoff, Sara Tolaney, and Ayca Gucalp
- Abstract
Supplementary Figure 1 - PDF file 44K, Supplementary Figure 1A-D. Median hormone levels at baseline, C2 and EOS
- Published
- 2023
- Full Text
- View/download PDF
29. Data from Functional Mapping of AKT Signaling and Biomarkers of Response from the FAIRLANE Trial of Neoadjuvant Ipatasertib plus Paclitaxel for Triple-Negative Breast Cancer
- Author
-
Steven J. Isakoff, Mafalda Oliveira, Emanuel F. Petricoin, Matthew J. Wongchenko, Patricia Villagrasa, Eva M. Ciruelos, Debra A. Pratt, Begoña Bermejo, Miguel J. Gil-Gil, José Luis Passos-Coelho, Jay Andersen, Isabel Calvo, Paolo G. Nuciforo, Cristina Saura, Rosa I. Gallagher, Malgorzata Nowicka, Julia Wulfkuhle, and Zhen Shi
- Abstract
Purpose:Despite extensive genomic and transcriptomic profiling, it remains unknown how signaling pathways are differentially activated and how tumors are differentially sensitized to certain perturbations. Here, we aim to characterize AKT signaling activity and its association with other genomic or IHC-based PI3K/AKT pathway biomarkers as well as the clinical activity of ipatasertib (AKT inhibitor) in the FAIRLANE trial.Experimental Design:In FAIRLANE, 151 patients with early triple-negative breast cancer (TNBC) were randomized 1:1 to receive paclitaxel with ipatasertib or placebo for 12 weeks prior to surgery. Adding ipatasertib did not increase pathologic complete response rate and numerically improved overall response rate by MRI. We used reverse-phase protein microarrays (RPPA) to examine the total level and/or phosphorylation states of over 100 proteins in various signaling or cell processes including PI3K/AKT and mTOR signaling. One hundred and twenty-five baseline and 127 on-treatment samples were evaluable by RPPA, with 110 paired samples at both time points.Results:Tumors with genomic/protein alterations in PIK3CA/AKT1/PTEN were associated with higher levels of AKT phosphorylation. In addition, phosphorylated AKT (pAKT) levels exhibited a significant association with enriched clinical benefit of ipatasertib, and identified patients who received benefit in the absence of PIK3CA/AKT1/PTEN alterations. Ipatasertib treatment led to a downregulation of AKT/mTORC1 signaling, which was more pronounced among the tumors with PIK3CA/AKT1/PTEN alterations or among the responders to the treatment.Conclusions:We showed that the high baseline pAKT levels are associated with the alterations of PI3K/AKT pathway components and enriched benefit of ipatasertib in TNBC.
- Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Tables from Identification of Somatically Acquired BRCA1/2 Mutations by cfDNA Analysis in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Daniel A. Haber, Shyamala Maheswaran, Leif W. Ellisen, Lee Zou, Gad Getz, A. John Iafrate, Mehmet Toner, Richard Lanman, Jerry Younger, Kristen Shannon, Beverly Moy, Laura Spring, Seth Wander, Douglas Micalizzi, Dejan Juric, Steven J. Isakoff, Jochen Lennerz, Giuliana Malvarosa, Brittany A. Reeves, Brian Chirn, Whijae Roh, Becky Nagy, Erica Blouch, Andrzej Niemierko, Antoine Simoneau, Michael S. Lawrence, Taronish Dubash, and Neelima Vidula
- Abstract
Supplemental tables
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Table S1 from Predicting Responses to Neoadjuvant Chemotherapy in Breast Cancer: ACRIN 6691 Trial of Diffuse Optical Spectroscopic Imaging
- Author
-
David Mankoff, Steven J. Isakoff, Stefan A. Carp, David A. Boas, Nola Hylton, Bradley S. Snyder, Mitchell Schnall, So Hyun Chung, Arjun G. Yodh, Peter A. Kaufman, Shudong Jiang, Brian W. Pogue, Keith D. Paulsen, Wei Yang, Darren Roblyer, Rita S. Mehta, Philip M. Carpenter, Albert E. Cerussi, Thomas D. O'Sullivan, Anaïs Leproux, Zheng Zhang, and Bruce J. Tromberg
- Abstract
Accrual table by institution.
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figures from Expanded Genomic Profiling of Circulating Tumor Cells in Metastatic Breast Cancer Patients to Assess Biomarker Status and Biology Over Time (CALGB 40502 and CALGB 40503, Alliance)
- Author
-
John W. Park, Laura van't Veer, Maura Dickler, Steven J. Isakoff, William T. Barry, Terry Hyslop, Brandelyn Pitcher, Jin Sun Lee, Janet H. Scott, Eduardo V. Sosa, Praveen Pendyala, Ritu Roy, Louai Hauranieh, Denise M. Wolf, Hope S. Rugo, and Mark Jesus M. Magbanua
- Abstract
Supplementary Figure 1. Development, optimization and performance of the gene expression assay. Supplementary Figure 2. Evaluation of assay performance using cancer cell line spike-in models. Supplementary Figure 3. Sample processing and quality control measures Supplementary Figure 4. Expression analysis of CTCs and matched normal blood samples. Supplementary Figure 5. Subtype and proliferation status. Supplementary Figure 6. Serial analysis. Supplementary Figure 7. Clustering analysis of copy number data. Supplementary Figure 8. ESR1 and ERBB2 status in CTCs and patient outcome Supplementary Figure 9. CTC molecular subtype and patient outcome.
- Published
- 2023
- Full Text
- View/download PDF
33. Data from Predicting Responses to Neoadjuvant Chemotherapy in Breast Cancer: ACRIN 6691 Trial of Diffuse Optical Spectroscopic Imaging
- Author
-
David Mankoff, Steven J. Isakoff, Stefan A. Carp, David A. Boas, Nola Hylton, Bradley S. Snyder, Mitchell Schnall, So Hyun Chung, Arjun G. Yodh, Peter A. Kaufman, Shudong Jiang, Brian W. Pogue, Keith D. Paulsen, Wei Yang, Darren Roblyer, Rita S. Mehta, Philip M. Carpenter, Albert E. Cerussi, Thomas D. O'Sullivan, Anaïs Leproux, Zheng Zhang, and Bruce J. Tromberg
- Abstract
The prospective multicenter ACRIN 6691 trial was designed to evaluate whether changes from baseline to mid-therapy in a diffuse optical spectroscopic imaging (DOSI)–derived imaging endpoint, the tissue optical index (TOI), predict pathologic complete response (pCR) in women undergoing breast cancer neoadjuvant chemotherapy (NAC). DOSI instruments were constructed at the University of California, Irvine (Irvine, CA), and delivered to six institutions where 60 subjects with newly diagnosed breast tumors (at least 2 cm in the longest dimension) were enrolled over a 2-year period. Bedside DOSI images of the tissue concentrations of deoxy-hemoglobin (ctHHb), oxy-hemoglobin (ctHbO2), water (ctH2O), lipid, and TOI (ctHHb × ctH2O/lipid) were acquired on both breasts up to four times during NAC treatment: baseline, 1-week, mid-point, and completion. Of the 34 subjects (mean age 48.4 ± 10.7 years) with complete, evaluable data from both normal and tumor-containing breast, 10 (29%) achieved pCR as determined by central pathology review. The percent change in tumor-to-normal TOI ratio (%TOITN) from baseline to mid-therapy ranged from −82% to 321%, with a median of −36%. Using pCR as the reference standard and ROC curve methodology, %TOITN AUC was 0.60 (95% CI, 0.39–0.81). In the cohort of 17 patients with baseline tumor oxygen saturation (%StO2) greater than the 77% population median, %TOITN AUC improved to 0.83 (95% CI, 0.63–1.00). We conclude that the combination of baseline functional properties and dynamic optical response shows promise for clinical outcome prediction. Cancer Res; 76(20); 5933–44. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Methods, Supplementary Figures 1-4, Supplementary Tables 1-6 from Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer
- Author
-
Andrea L. Richardson, Daniel P. Silver, James M. Ford, Judy E. Garber, Anne-Renee Hartman, Jerry S. Lanchbury, Joshua T. Jones, Victor Abkevich, Chris Neff, Diana Iliev, Zaina Sangale, Alexander Gutin, April Greene-Colozzi, Paula D. Ryan, Steven J. Isakoff, Nadine M. Tung, Eric P. Winer, William T. Barry, Zoltan Szallasi, Kristin C. Jensen, Gordon B. Mills, Bryan Hennessy, Julia Reid, Kirsten M. Timms, and Melinda L. Telli
- Abstract
Figure S1. Summary of HRD testing in PrECOG and combined cisplatin cohorts; Figure S2. Distribution of HRD scores in PrECOG and combined cisplatin cohorts; Figure S3. Performance of HRD in predicting response in PrECOG and combined cisplatin cohorts; Figure S4. Logistic regression models to predict response in PrECOG and combined cisplatin cohorts; Table S1. Study cohorts for threshold training set; Table S2. Summary statistics of HRD scores in BRCA1/2 deficient samples by tissue in training; Table S3. Patient clinical and demographic data; Table S4. HRD score and association with therapy response in entire cohorts; Table S5. Univariate associations between clinical variables and RCB 0/1 or HR deficiency status; Table S6. Analysis of RCB 0/1 and pCR in combined PrECOG 0105 and Cisplatin Trials cohorts (logistic regression models adjusted for cohort)
- Published
- 2023
- Full Text
- View/download PDF
35. Supplementary Figure S3 from Identification of Somatically Acquired BRCA1/2 Mutations by cfDNA Analysis in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Daniel A. Haber, Shyamala Maheswaran, Leif W. Ellisen, Lee Zou, Gad Getz, A. John Iafrate, Mehmet Toner, Richard Lanman, Jerry Younger, Kristen Shannon, Beverly Moy, Laura Spring, Seth Wander, Douglas Micalizzi, Dejan Juric, Steven J. Isakoff, Jochen Lennerz, Giuliana Malvarosa, Brittany A. Reeves, Brian Chirn, Whijae Roh, Becky Nagy, Erica Blouch, Andrzej Niemierko, Antoine Simoneau, Michael S. Lawrence, Taronish Dubash, and Neelima Vidula
- Abstract
MAF of mutations detected
- Published
- 2023
- Full Text
- View/download PDF
36. Data from Expanded Genomic Profiling of Circulating Tumor Cells in Metastatic Breast Cancer Patients to Assess Biomarker Status and Biology Over Time (CALGB 40502 and CALGB 40503, Alliance)
- Author
-
John W. Park, Laura van't Veer, Maura Dickler, Steven J. Isakoff, William T. Barry, Terry Hyslop, Brandelyn Pitcher, Jin Sun Lee, Janet H. Scott, Eduardo V. Sosa, Praveen Pendyala, Ritu Roy, Louai Hauranieh, Denise M. Wolf, Hope S. Rugo, and Mark Jesus M. Magbanua
- Abstract
Purpose: We profiled circulating tumor cells (CTCs) to study the biology of blood-borne metastasis and to monitor biomarker status in metastatic breast cancer (MBC).Methods: CTCs were isolated from 105 patients with MBC using EPCAM-based immunomagnetic enrichment and fluorescence-activated cells sorting (IE/FACS), 28 of whom had serial CTC analysis (74 samples, 2–5 time points). CTCs were subjected to microfluidic-based multiplex QPCR array of 64 cancer-related genes (n = 151) and genome-wide copy-number analysis by array comparative genomic hybridization (aCGH; n = 49).Results: Combined transcriptional and genomic profiling showed that CTCs were 26% ESR1−ERBB2−, 48% ESR1+ERBB2−, and 27% ERBB2+. Serial testing showed that ERBB2 status was more stable over time compared with ESR1 and proliferation (MKI67) status. While cell-to-cell heterogeneity was observed at the single-cell level, with increasingly stable expression in larger pools, patient-specific CTC expression “fingerprints” were also observed. CTC copy-number profiles clustered into three groups based on the extent of genomic aberrations and the presence of large chromosomal imbalances. Comparative analysis showed discordance in ESR1/ER (27%) and ERBB2/HER2 (23%) status between CTCs and matched primary tumors. CTCs in 65% of the patients were considered to have low proliferation potential. Patients who harbored CTCs with high proliferation (MKI67) status had significantly reduced progression-free survival (P = 0.0011) and overall survival (P = 0.0095) compared with patients with low proliferative CTCs.Conclusions: We demonstrate an approach for complete isolation of EPCAM-positive CTCs and downstream comprehensive transcriptional/genomic characterization to examine the biology and assess breast cancer biomarkers in these cells over time. Clin Cancer Res; 24(6); 1486–99. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
37. Data from Identification of Somatically Acquired BRCA1/2 Mutations by cfDNA Analysis in Patients with Metastatic Breast Cancer
- Author
-
Aditya Bardia, Daniel A. Haber, Shyamala Maheswaran, Leif W. Ellisen, Lee Zou, Gad Getz, A. John Iafrate, Mehmet Toner, Richard Lanman, Jerry Younger, Kristen Shannon, Beverly Moy, Laura Spring, Seth Wander, Douglas Micalizzi, Dejan Juric, Steven J. Isakoff, Jochen Lennerz, Giuliana Malvarosa, Brittany A. Reeves, Brian Chirn, Whijae Roh, Becky Nagy, Erica Blouch, Andrzej Niemierko, Antoine Simoneau, Michael S. Lawrence, Taronish Dubash, and Neelima Vidula
- Abstract
Purpose:Plasma genotyping may identify mutations in potentially “actionable” cancer genes, such as BRCA1/2, but their clinical significance is not well-defined. We evaluated the characteristics of somatically acquired BRCA1/2 mutations in patients with metastatic breast cancer (MBC).Experimental Design:Patients with MBC undergoing routine cell-free DNA (cfDNA) next-generation sequencing (73-gene panel) before starting a new therapy were included. Somatic BRCA1/2 mutations were classified as known germline pathogenic mutations or novel variants, and linked to clinicopathologic characteristics. The effect of the PARP inhibitor, olaparib, was assessed in vitro, using cultured circulating tumor cells (CTCs) from a patient with a somatically acquired BRCA1 mutation and a second patient with an acquired BRCA2 mutation.Results:Among 215 patients with MBC, 29 (13.5%) had somatic cfDNA BRCA1/2 mutations [nine (4%) known germline pathogenic and rest (9%) novel variants]. Known germline pathogenic BRCA1/2 mutations were common in younger patients (P = 0.008), those with triple-negative disease (P = 0.022), and they were more likely to be protein-truncating alterations and be associated with TP53 mutations. Functional analysis of a CTC culture harboring a somatic BRCA1 mutation demonstrated high sensitivity to PARP inhibition, while another CTC culture harboring a somatic BRCA2 mutation showed no differential sensitivity. Across the entire cohort, APOBEC mutational signatures (COSMIC Signatures 2 and 13) and the “BRCA” mutational signature (COSMIC Signature 3) were present in BRCA1/2-mutant and wild-type cases, demonstrating the high mutational burden associated with advanced MBC.Conclusions:Somatic BRCA1/2 mutations are readily detectable in MBC by cfDNA analysis, and may be present as both known germline pathogenic and novel variants.
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Legend from Phase II Trial of Bicalutamide in Patients with Androgen Receptor–Positive, Estrogen Receptor–Negative Metastatic Breast Cancer
- Author
-
Tiffany A. Traina, Clifford A. Hudis, Kimberly N. Feigin, Sujata Patil, Dilip D. Giri, Arooj Akhtar, Joseph M. Gonzalez, Lamia F. Momen, Mary Ellen Moynahan, Michael Danso, Ashley S. Doane, Vered Stearns, Andres Forero, Lisle Nabell, Hope Rugo, Kimberly Blackwell, Lisa A. Carey, Minetta C. Liu, James N. Ingle, Steven J. Isakoff, Sara Tolaney, and Ayca Gucalp
- Abstract
Supplementary Legend - PDF file 22K, Supplementary Legend
- Published
- 2023
- Full Text
- View/download PDF
39. Data from Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer
- Author
-
Andrea L. Richardson, Daniel P. Silver, James M. Ford, Judy E. Garber, Anne-Renee Hartman, Jerry S. Lanchbury, Joshua T. Jones, Victor Abkevich, Chris Neff, Diana Iliev, Zaina Sangale, Alexander Gutin, April Greene-Colozzi, Paula D. Ryan, Steven J. Isakoff, Nadine M. Tung, Eric P. Winer, William T. Barry, Zoltan Szallasi, Kristin C. Jensen, Gordon B. Mills, Bryan Hennessy, Julia Reid, Kirsten M. Timms, and Melinda L. Telli
- Abstract
Purpose:BRCA1/2-mutated and some sporadic triple-negative breast cancers (TNBC) have DNA repair defects and are sensitive to DNA-damaging therapeutics. Recently, three independent DNA-based measures of genomic instability were developed on the basis of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST).Experimental Design: We assessed a combined homologous recombination deficiency (HRD) score, an unweighted sum of LOH, TAI, and LST scores, in three neoadjuvant TNBC trials of platinum-containing therapy. We then tested the association of HR deficiency, defined as HRD score ≥42 or BRCA1/2 mutation, with response to platinum-based therapy.Results: In a trial of neoadjuvant platinum, gemcitabine, and iniparib, HR deficiency predicted residual cancer burden score of 0 or I (RCB 0/I) and pathologic complete response (pCR; OR = 4.96, P = 0.0036; OR = 6.52, P = 0.0058). HR deficiency remained a significant predictor of RCB 0/I when adjusted for clinical variables (OR = 5.86, P = 0.012). In two other trials of neoadjuvant cisplatin therapy, HR deficiency predicted RCB 0/I and pCR (OR = 10.18, P = 0.0011; OR = 17.00, P = 0.0066). In a multivariable model of RCB 0/I, HR deficiency retained significance when clinical variables were included (OR = 12.08, P = 0.0017). When restricted to BRCA1/2 nonmutated tumors, response was higher in patients with high HRD scores: RCB 0/I P = 0.062, pCR P = 0.063 in the neoadjuvant platinum, gemcitabine, and iniparib trial; RCB 0/I P = 0.0039, pCR P = 0.018 in the neoadjuvant cisplatin trials.Conclusions: HR deficiency identifies TNBC tumors, including BRCA1/2 nonmutated tumors more likely to respond to platinum-containing therapy. Clin Cancer Res; 22(15); 3764–73. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Figure 2 from Phase II Trial of Bicalutamide in Patients with Androgen Receptor–Positive, Estrogen Receptor–Negative Metastatic Breast Cancer
- Author
-
Tiffany A. Traina, Clifford A. Hudis, Kimberly N. Feigin, Sujata Patil, Dilip D. Giri, Arooj Akhtar, Joseph M. Gonzalez, Lamia F. Momen, Mary Ellen Moynahan, Michael Danso, Ashley S. Doane, Vered Stearns, Andres Forero, Lisle Nabell, Hope Rugo, Kimberly Blackwell, Lisa A. Carey, Minetta C. Liu, James N. Ingle, Steven J. Isakoff, Sara Tolaney, and Ayca Gucalp
- Abstract
Supplementary Figure 2 - PDF file 275K, Supplementary Figure 2A-D. Changes in hormone levels over time per patient; responders (stable disease >6 months) highlighted in red
- Published
- 2023
- Full Text
- View/download PDF
41. 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
42. 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
43. Supplementary Data from Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis
- Author
-
Aditya Bardia, Lorenzo Trippa, Giovanni Parmigiani, Steven J. Isakoff, Beverly Moy, Brian Alexander, Barbara L. Smith, Kerry L. Reynolds, Rachel Greenup, Chandni Sharma, Andrea Arfe, Geoffrey Fell, and Laura M. Spring
- Abstract
Online supplement
- Published
- 2023
- Full Text
- View/download PDF
44. Data from Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis
- Author
-
Aditya Bardia, Lorenzo Trippa, Giovanni Parmigiani, Steven J. Isakoff, Beverly Moy, Brian Alexander, Barbara L. Smith, Kerry L. Reynolds, Rachel Greenup, Chandni Sharma, Andrea Arfe, Geoffrey Fell, and Laura M. Spring
- Abstract
Purpose:While various studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy (NAT), the impact of additional adjuvant therapy after pCR is not known.Experimental Design:PubMed was searched for studies with NAT for breast cancer and individual patient-level data was extracted for analysis using plot digitizer software. HRs, with 95% probability intervals (PI), measuring the association between pCR and overall survival (OS) or event-free survival (EFS), were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models including pCR as predictor.Results:Overall, 52 of 3,209 publications met inclusion criteria, totaling 27,895 patients. Patients with a pCR after NAT had significantly better EFS (HR = 0.31; 95% PI, 0.24–0.39), particularly for triple-negative (HR = 0.18; 95% PI, 0.10–0.31) and HER2+ (HR = 0.32; 95% PI, 0.21–0.47) disease. Similarly, pCR after NAT was also associated with improved survival (HR = 0.22; 95% PI, 0.15–0.30). The association of pCR with improved EFS was similar among patients who received subsequent adjuvant chemotherapy (HR = 0.36; 95% PI, 0.19–0.67) and those without adjuvant chemotherapy (HR = 0.36; 95% PI, 0.27–0.54), with no significant difference between the two groups (P = 0.60).Conclusions:Achieving pCR following NAT is associated with significantly better EFS and OS, particularly for triple-negative and HER2+ breast cancer. The similar outcomes with or without adjuvant chemotherapy in patients who attain pCR likely reflects tumor biology and systemic clearance of micrometastatic disease, highlighting the potential of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response.See related commentary by Esserman, p. 2771
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Table from Functional Mapping of AKT Signaling and Biomarkers of Response from the FAIRLANE Trial of Neoadjuvant Ipatasertib plus Paclitaxel for Triple-Negative Breast Cancer
- Author
-
Steven J. Isakoff, Mafalda Oliveira, Emanuel F. Petricoin, Matthew J. Wongchenko, Patricia Villagrasa, Eva M. Ciruelos, Debra A. Pratt, Begoña Bermejo, Miguel J. Gil-Gil, José Luis Passos-Coelho, Jay Andersen, Isabel Calvo, Paolo G. Nuciforo, Cristina Saura, Rosa I. Gallagher, Malgorzata Nowicka, Julia Wulfkuhle, and Zhen Shi
- Abstract
Supplementary Table from Functional Mapping of AKT Signaling and Biomarkers of Response from the FAIRLANE Trial of Neoadjuvant Ipatasertib plus Paclitaxel for Triple-Negative Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
46. Supplementary Methods from Predicting Responses to Neoadjuvant Chemotherapy in Breast Cancer: ACRIN 6691 Trial of Diffuse Optical Spectroscopic Imaging
- Author
-
David Mankoff, Steven J. Isakoff, Stefan A. Carp, David A. Boas, Nola Hylton, Bradley S. Snyder, Mitchell Schnall, So Hyun Chung, Arjun G. Yodh, Peter A. Kaufman, Shudong Jiang, Brian W. Pogue, Keith D. Paulsen, Wei Yang, Darren Roblyer, Rita S. Mehta, Philip M. Carpenter, Albert E. Cerussi, Thomas D. O'Sullivan, Anaïs Leproux, Zheng Zhang, and Bruce J. Tromberg
- Abstract
List of Chemotherapy regimens used in this study.
- Published
- 2023
- Full Text
- View/download PDF
47. Supplementary Figure from Functional Mapping of AKT Signaling and Biomarkers of Response from the FAIRLANE Trial of Neoadjuvant Ipatasertib plus Paclitaxel for Triple-Negative Breast Cancer
- Author
-
Steven J. Isakoff, Mafalda Oliveira, Emanuel F. Petricoin, Matthew J. Wongchenko, Patricia Villagrasa, Eva M. Ciruelos, Debra A. Pratt, Begoña Bermejo, Miguel J. Gil-Gil, José Luis Passos-Coelho, Jay Andersen, Isabel Calvo, Paolo G. Nuciforo, Cristina Saura, Rosa I. Gallagher, Malgorzata Nowicka, Julia Wulfkuhle, and Zhen Shi
- Abstract
Supplementary Figure from Functional Mapping of AKT Signaling and Biomarkers of Response from the FAIRLANE Trial of Neoadjuvant Ipatasertib plus Paclitaxel for Triple-Negative Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary Table 1 from Phase II Trial of Bicalutamide in Patients with Androgen Receptor–Positive, Estrogen Receptor–Negative Metastatic Breast Cancer
- Author
-
Tiffany A. Traina, Clifford A. Hudis, Kimberly N. Feigin, Sujata Patil, Dilip D. Giri, Arooj Akhtar, Joseph M. Gonzalez, Lamia F. Momen, Mary Ellen Moynahan, Michael Danso, Ashley S. Doane, Vered Stearns, Andres Forero, Lisle Nabell, Hope Rugo, Kimberly Blackwell, Lisa A. Carey, Minetta C. Liu, James N. Ingle, Steven J. Isakoff, Sara Tolaney, and Ayca Gucalp
- Abstract
Supplementary Table 1 - PDF file 4K, Supplementary Table 1. Median hormone levels at baseline, start of cycle 2 (C2), and end of study (EOS) and the percent change in levels from baseline to C2 and C2 to EOS
- Published
- 2023
- Full Text
- View/download PDF
49. Early prediction of neoadjuvant chemotherapy outcome with longitudinal breast diffuse optical tomography (Conference Presentation)
- Author
-
Bin Deng, Ailis Muldoon, Jayne Cormier, Mansi A. Saksena, Steven J. Isakoff, and Stefan A. Carp
- Published
- 2023
- Full Text
- View/download PDF
50. Stressed target cancer cells drive nongenetic reprogramming of CAR T cells and tumor microenvironment, overcoming multiple obstacles of CAR T therapy for solid tumors
- Author
-
Yufeng Wang, David L. Drum, Ruochuan Sun, Yida Zhang, Ling Yu, Lin Jia, Steven J Isakoff, Allison M Kehlmann, Ali Emre Dal, Gianpietro Dotti, Hui Zheng, Cristina R Ferrone, Alphonse G Taghian, Albert B DeLeo, Hanyu Zhang, Youssef Jounaidi, Song Fan, Peigen Huang, Cheng Wang, Jibing Yang, Genevieve M Boland, Ruslan I Sadreyev, LaiPing Wong, Soldano Ferrone, and Xinhui Wang
- Abstract
The poor efficacy of chimeric antigen receptor T-cell therapy (CAR T) for solid tumor is due to insufficient CAR T cell tumor infiltration, in vivo expansion, persistence, and effector function, as well as exhaustion, intrinsic target antigen heterogeneity or antigen loss of target cancer cells, and immunosuppressive tumor microenvironment (TME). Here we describe a broadly applicable nongenetic approach that simultaneously addresses the multiple challenges of CAR T as a therapy for solid tumors. The approach massively reprograms CAR T cells by exposing them to stressed target cancer cells which have been exposed to the cell stress inducer disulfiram (DSF) and copper (Cu)(DSF/Cu) plus ionizing irradiation (IR). The reprogrammed CAR T cells acquired early memory-like characteristics, potent cytotoxicity, enhanced in vivo expansion, persistence, and decreased exhaustion. Tumors stressed by DSF/Cu and IR also reprogrammed and reversed immunosuppressive TME in humanized mice. The reprogrammed CAR T cells, derived from peripheral blood mononuclear cells (PBMC) of healthy or metastatic breast cancer patients, induced robust, sustained memory and curative anti-solid tumor responses in multiple xenograft mouse models, establishing proof of concept for empowering CAR T by stressing tumor as a novel therapy for solid tumor.
- 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.