40 results on '"Pamela Munster"'
Search Results
2. Immunotherapy in breast cancer: A clinician's perspective
- Author
-
Sibapriya Chaudhuri, Scott Thomas, and Pamela Munster
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Globally over 2 million women are diagnosed with breast cancer each year despite major advances in detection and treatment of the disease. Breast cancer is comprised of several distinct subtypes and understanding the heterogeneity of the disease has become crucial for treatment planning. Therapeutic strategies span from a hormone therapy-based focus for women with estrogen receptor positive breast cancer to targeting human epidermal growth factor (HER2) by small molecules, antibody-drug-conjugates (ADC) and monoclonal antibodies in those with HER2 overexpression. Other novel treatment strategies for select subgroups of patients include the cyclin-dependent kinase 4/6 (CDK4/6) inhibitors for women with estrogen receptor positive tumors, the poly ADP ribose polymerase (PARP) inhibitors for those with BRCA mutations, and phosphoinositide 3-kinase (PI3K) inhibitors for women with tumors harboring phophatidylinositol-4,5-bisphosphate 3 kinase catalytic subunit alpha (PIK3CA) mutations. In contrast, the treatment for women with triple negative breast cancer has until recently been solely limited to chemotherapy. The profound impact of immunotherapy on cancer treatment in general has created much hope for its potential in breast cancer. This review will focus on the current advances and the research of immunotherapy in breast cancer, particularly on immune checkpoint inhibitors, adoptive cell transfer and cancer vaccines.
- Published
- 2021
- Full Text
- View/download PDF
3. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer
- Author
-
Anniina Färkkilä, Doga C. Gulhan, Julia Casado, Connor A. Jacobson, Huy Nguyen, Bose Kochupurakkal, Zoltan Maliga, Clarence Yapp, Yu-An Chen, Denis Schapiro, Yinghui Zhou, Julie R. Graham, Bruce J. Dezube, Pamela Munster, Sandro Santagata, Elizabeth Garcia, Scott Rodig, Ana Lako, Dipanjan Chowdhury, Geoffrey I. Shapiro, Ursula A. Matulonis, Peter J. Park, Sampsa Hautaniemi, Peter K. Sorger, Elizabeth M. Swisher, Alan D. D’Andrea, and Panagiotis A. Konstantinopoulos
- Subjects
Science - Abstract
A Phase I/II trial previously revealed variable anti-tumor efficacy of the PARP inhibitor niraparib in combination with the PD-1 inhibitor pembrolizumab in platinum-resistant ovarian cancer patients. Here, the authors perform an integrated genomic and immunomics analysis of tumor samples from the same patients and find potential predictive biomarkers of response to such combination therapy.
- Published
- 2020
- Full Text
- View/download PDF
4. Safety and clinical activity of intratumoral MEDI9197 alone and in combination with durvalumab and/or palliative radiation therapy in patients with advanced solid tumors
- Author
-
Christopher Morehouse, Rakesh Kumar, Antoine Hollebecque, Aurélien Marabelle, Zachary A Cooper, Joshua Brody, Farzana Walcott, Charles Ferte, Shilpa Gupta, David S Hong, Lillian Siu, Antonio Jimeno, Pamela Munster, Juneko Grilley-Olson, Alain H Rook, Rebecca K S Wong, James W Welsh, Yuling Wu, and Oday Hamid
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background MEDI9197 is an intratumorally administered toll-like receptor 7 and 8 agonist. In mice, MEDI9197 modulated antitumor immune responses, inhibited tumor growth and increased survival. This first-time-in-human, phase 1 study evaluated MEDI9197 with or without the programmed cell death ligand-1 (PD-L1) inhibitor durvalumab and/or palliative radiation therapy (RT) for advanced solid tumors.Patients and methods Eligible patients had at least one cutaneous, subcutaneous, or deep-seated lesion suitable for intratumoral (IT) injection. Dose escalation used a standard 3+3 design. Patients received IT MEDI9197 0.005–0.055 mg with or without RT (part 1), or IT MEDI9197 0.005 or 0.012 mg plus durvalumab 1500 mg intravenous with or without RT (part 3), in 4-week cycles. Primary endpoints were safety and tolerability. Secondary endpoints included pharmacokinetics, pharmacodynamics, and objective response based on Response Evaluation Criteria for Solid Tumors version 1.1. Exploratory endpoints included tumor and peripheral biomarkers that correlate with biological activity or predict response.Results From November 2015 to March 2018, part 1 enrolled 35 patients and part 3 enrolled 17 patients; five in part 1 and 2 in part 3 received RT. The maximum tolerated dose of MEDI9197 monotherapy was 0.037 mg, with dose-limiting toxicity (DLT) of cytokine release syndrome in two patients (one grade 3, one grade 4) and 0.012 mg in combination with durvalumab 1500 mg with DLT of MEDI9197-related hemorrhagic shock in one patient (grade 5) following liver metastasis rupture after two cycles of MEDI9197. Across parts 1 and 3, the most frequent MEDI9197-related adverse events (AEs) of any grade were fever (56%), fatigue (31%), and nausea (21%). The most frequent MEDI9197-related grade ≥3 events were decreased lymphocytes (15%), neutrophils (10%), and white cell counts (10%). MEDI9197 increased tumoral CD8+ and PD-L1+ cells, inducing type 1 and 2 interferons and Th1 response. There were no objective clinical responses; 10 patients in part 1 and 3 patients in part 3 had stable disease ≥8 weeks.Conclusion IT MEDI9197 was feasible for subcutaneous/cutaneous lesions but AEs precluded its use in deep-seated lesions. Although no patients responded, MEDI9197 induced systemic and intratumoral immune activation, indicating potential value in combination regimens in other patient populations.Trial registration number NCT02556463.
- Published
- 2020
- Full Text
- View/download PDF
5. Author Correction: Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer
- Author
-
Anniina Färkkilä, Doga C. Gulhan, Julia Casado, Connor A. Jacobson, Huy Nguyen, Bose Kochupurakkal, Zoltan Maliga, Clarence Yapp, Yu-An Chen, Denis Schapiro, Yinghui Zhou, Julie R. Graham, Bruce J. Dezube, Pamela Munster, Sandro Santagata, Elizabeth Garcia, Scott Rodig, Ana Lako, Dipanjan Chowdhury, Geoffrey I. Shapiro, Ursula A. Matulonis, Peter J. Park, Sampsa Hautaniemi, Peter K. Sorger, Elizabeth M. Swisher, Alan D. D’Andrea, and Panagiotis A. Konstantinopoulos
- Subjects
Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
6. Phase II Study of Enzalutamide for Patients With Androgen Receptor–Positive Salivary Gland Cancers (Alliance A091404)
- Author
-
Alan L. Ho, Nathan R. Foster, Alex J. Zoroufy, Jordan D. Campbell, Francis Worden, Katharine Price, Douglas Adkins, Daniel W. Bowles, Hyunseok Kang, Barbara Burtness, Eric Sherman, Roscoe Morton, Luc G.T. Morris, Zaineb Nadeem, Nora Katabi, Pamela Munster, and Gary K. Schwartz
- Subjects
Male ,Cancer Research ,Oncology ,Receptors, Androgen ,Nitriles ,Phenylthiohydantoin ,Humans ,Female ,Androgen Antagonists ,Salivary Gland Neoplasms - Abstract
PURPOSE The androgen receptor (AR) is expressed (+) in a subset of salivary gland cancers (SGCs). This phase II trial evaluated the efficacy of the antiandrogen enzalutamide in AR+ SGC. METHODS Patients with locally advanced/unresectable or metastatic AR+ SGCs were enrolled. Enzalutamide (160 mg) was given orally once daily. The primary end point was the best overall response rate per RECIST v1.1 within eight cycles. Confirmed responses in ≥ 5 of 41 patients would be considered promising. Secondary end points were progression-free survival, overall survival, and safety. RESULTS Forty-six patients were enrolled; 30 (65.2%) received prior systemic therapy, including 13 (28.3%) with AR-targeted drugs. Of seven (15.2%) partial responses (PRs), only two (4.3%) were confirmed per protocol and counted toward the primary end point. Twenty-four patients (52.2%) had stable disease; 15 (32.6%) had progression of disease as best response. Twenty-six patients (56.5%) experienced tumor regression in target lesions; 18 (39.1%) had partial response/stable disease ≥ 6 months. Tumor regressions were observed in female patients (5 of 6 [83.3%]) and those who received prior AR– (6 of 13 [46.2%]) or human epidermal growth factor receptor 2–targeted therapies (5 of 8 [62.5%]). Three patients remained on treatment at data cutoff (duration, 32.2-49.8 months). The median progression-free survival was 5.6 months (95% CI, 3.7 to 7.5); the median overall survival was 17.0 months (95% CI, 11.8 to 30.0). The most common adverse events were fatigue, hypertension, hot flashes, and weight loss. Total and free testosterone levels increased by a mean of 61.2% and 48.8%, respectively, after enzalutamide. CONCLUSION Enzalutamide demonstrated limited activity in AR+ SGC, failing to meet protocol-defined success in part because of a lack of response durability. Strategies to enhance the efficacy of antiandrogen therapy are needed.
- Published
- 2022
7. Data from Complement Activation and Intraventricular Rituximab Distribution in Recurrent Central Nervous System Lymphoma
- Author
-
James L. Rubenstein, Marc A. Shuman, Clifford A. Lowell, Pamela Munster, Soonmee Cha, Lingjing Chen, Valerie S. Wong, Jing Li, and Cigall Kadoch
- Abstract
Purpose: To elucidate the mechanistic basis for efficacy of intrathecal rituximab. We evaluated complement activation as well as the pharmacokinetics of intraventricular rituximab in patients who participated in two phase 1 multicenter studies.Experimental Design: We evaluated complement activation as a candidate mediator of rituximab within the central nervous system (CNS). Complement C3 and C5b-9 were quantified by ELISA in serial cerebrospinal fluid (CSF) specimens after intraventricular rituximab administration. We determined rituximab concentration profiles in CSF and serum. A population three- compartment pharmacokinetic model was built to describe the disposition of rituximab following intraventricular administration. The model was derived from results of the first trial and validated with results of the second trial.Results: Complement C3 and C5b-9 were reproducibly activated in CSF after intraventricular rituximab. Ectopic expression of C3 mRNA and protein within CNS lymphoma lesions was localized to myeloid cells. Constitutive high C3 activation at baseline was associated with adverse prognosis. A pharmacokinetic model was built, which contains three distinct compartments, to describe the distribution of rituximab within the neuroaxis after intraventricular administration.Conclusions: We provide the first evidence of C3 activation within the neuroaxis with intraventricular immunotherapy and suggest that complement may contribute to immunotherapeutic responses of rituximab in CNS lymphoma. Penetration of rituximab into neural tissue is supported by this pharmacokinetic model and may contribute to efficacy. These findings have general implications for intraventricular immunotherapy. Our data highlight potential innovations to improve efficacy of intraventricular immunotherapy both via modulation of the innate immune response as well as innovations in drug delivery. Clin Cancer Res; 20(4); 1029–41. ©2013 AACR.
- Published
- 2023
8. Data from First-in-Human Phase I Study of GSK2126458, an Oral Pan-Class I Phosphatidylinositol-3-Kinase Inhibitor, in Patients with Advanced Solid Tumor Malignancies
- Author
-
Razelle Kurzrock, Shannon R. Morris, Thomas A. Lampkin, Deborah A. Smith, Laurel Adams, Michael Durante, Joseph F. Kleha, Neeraj Agarwal, Emily Bergsland, E. Claire Dees, Theresa L. Werner, Petronella O. Witteveen, Jennifer Specht, Ruud van der Noll, Jan H.M. Schellens, David Hong, Rahul Aggarwal, and Pamela Munster
- Abstract
Purpose: GSK2126458 (GSK458) is a potent inhibitor of PI3K (α, β, γ, and δ), with preclinical studies demonstrating broad antitumor activity. We performed a first-in-human phase I study in patients with advanced solid tumors.Materials and Methods: Patients received oral GSK458 once or twice daily in a dose-escalation design to define the maximum tolerated dose (MTD). Expansion cohorts evaluated pharmacodynamics, pharmacokinetics, and clinical activity in histologically and molecularly defined cohorts.Results: One hundred and seventy patients received doses ranging from 0.1 to 3 mg once or twice daily. Dose-limiting toxicities (grade 3 diarrhea, n = 4; fatigue and rash, n = 1) occurred in 5 patients (n = 3 at 3 mg/day). The MTD was 2.5 mg/day (MTD with twice daily dosing undefined). The most common grade ≥3 treatment-related adverse events included diarrhea (8%) and skin rash (5%). Pharmacokinetic analyses demonstrated increased duration of drug exposure above target level with twice daily dosing. Fasting insulin and glucose levels increased with dose and exposure of GSK458. Durable objective responses (ORs) were observed across multiple tumor types (sarcoma, kidney, breast, endometrial, oropharyngeal, and bladder cancer). Responses were not associated with PIK3CA mutations (OR rate: 5% wild-type vs. 6% mutant).Conclusions: Although the MTD of GSK458 was 2.5 mg once daily, twice-daily dosing may increase duration of target inhibition. Fasting insulin and glucose levels served as pharmacodynamic markers of drug exposure. Select patients achieved durable responses; however, PIK3CA mutations were neither necessary nor predictive of response. Combination treatment strategies and novel biomarkers may be needed to optimally target PI3K. Clin Cancer Res; 22(8); 1932–9. ©2015 AACR.
- Published
- 2023
9. Supplemental Figure 2 from Complement Activation and Intraventricular Rituximab Distribution in Recurrent Central Nervous System Lymphoma
- Author
-
James L. Rubenstein, Marc A. Shuman, Clifford A. Lowell, Pamela Munster, Soonmee Cha, Lingjing Chen, Valerie S. Wong, Jing Li, and Cigall Kadoch
- Abstract
Supplemental Figure 2. Effect of hypothetical brain tissue volume (V3) on rituximab disposition in CSF, serum and brain tissue after intraventricular administration. Figure 2A. V3 = 0 ml. Figure 2B. V3 = 320 ml. Figure 2C. V3 = 5000 ml.
- Published
- 2023
10. Supplementary Table 1 and 2 from First-in-Human Phase I Study of GSK2126458, an Oral Pan-Class I Phosphatidylinositol-3-Kinase Inhibitor, in Patients with Advanced Solid Tumor Malignancies
- Author
-
Razelle Kurzrock, Shannon R. Morris, Thomas A. Lampkin, Deborah A. Smith, Laurel Adams, Michael Durante, Joseph F. Kleha, Neeraj Agarwal, Emily Bergsland, E. Claire Dees, Theresa L. Werner, Petronella O. Witteveen, Jennifer Specht, Ruud van der Noll, Jan H.M. Schellens, David Hong, Rahul Aggarwal, and Pamela Munster
- Abstract
Supplementary Table 1. Pharmacokinetic Parameters on Day 1 Following Once daily and Twice Daily Administration of GSK458 Supplementary Table 2. Best Objective Tumor Response by Dose Level (supplementary)
- Published
- 2023
11. Supplementary Figure 1 from First-in-Human Phase I Study of GSK2126458, an Oral Pan-Class I Phosphatidylinositol-3-Kinase Inhibitor, in Patients with Advanced Solid Tumor Malignancies
- Author
-
Razelle Kurzrock, Shannon R. Morris, Thomas A. Lampkin, Deborah A. Smith, Laurel Adams, Michael Durante, Joseph F. Kleha, Neeraj Agarwal, Emily Bergsland, E. Claire Dees, Theresa L. Werner, Petronella O. Witteveen, Jennifer Specht, Ruud van der Noll, Jan H.M. Schellens, David Hong, Rahul Aggarwal, and Pamela Munster
- Abstract
Duration of therapy and best objective response in the expansion cohorts (Breast, Bladder, Renal, and Endometrial Cancer)
- Published
- 2023
12. Supplemental Figure 1 from Complement Activation and Intraventricular Rituximab Distribution in Recurrent Central Nervous System Lymphoma
- Author
-
James L. Rubenstein, Marc A. Shuman, Clifford A. Lowell, Pamela Munster, Soonmee Cha, Lingjing Chen, Valerie S. Wong, Jing Li, and Cigall Kadoch
- Abstract
Supplemental Figure 1. Elevated C3a des-arg concentration in CSF is associated with shorter progression-free survival in newly-diagnosed patients with CNS involvement of aggressive non-Hodgkin’s lymphoma (N=35). The survival for the seven patients with elevated C3a in CSF at diagnosis was significantly shorter than for the 28 patients with low CSF C3a concentration at diagnosis. (P60 and ECOG >1 were also associated with shorter progression-free survival (P
- Published
- 2023
13. Abstract P1-17-10: H3B-6545, a novel selective estrogen receptor covalent antagonist (SERCA), in estrogen receptor positive (ER+), human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer - A phase II study
- Author
-
Erika P Hamilton, Judy S Wang, Timothy Pluard, Aki Morikawa, E Claire Dees, Robert H Jones, Barbara Haley, Anne Armstrong, Adam L Cohen, Pamela Munster, Gail S Wright, Fadi Kayali, Lisa Cantagallo, Manav Korpal, Jenny Long, Jianjun Xiao, Benoit Destenaves, Lei Gao, Tarek Sahmoud, Antonio Gualberto, and Dejan Juric
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: H3B-6545, a novel Selective ERα Covalent Antagonist (SERCA), inactivates both and wild-type and mutant ERα by targeting cysteine 530 and enforcing antagonist conformation. H3B-6545 demonstrated preclinical high activity in breast cancer models with constitutively active ESR1 mutations (Furman C, SABCS 2020) and clinical activity in pretreated women with ER+ breast cancer (Hamilton EP, ASCO 2021). Patients and Methods: The primary objective of the phase II is to estimate the objective response rate (ORR), clinical benefit rate (CBR), progression-free survival (PFS), and secondary objectives include safety. Results: 94 pts were treated with 450 mg daily, the recommended phase II dose: 11 in the phase I and 83 in the phase II parts of the trial. Patients and tumor characteristics were presented previously (Hamilton EP, ASCO, 2021). As of March 31, 2021, grade (gr) 2 or higher adverse events (AE) reported in ≥10% were anemia (19%), nausea (17%), fatigue (16%), and diarrhea (12%). Laboratory gr 2 or higher abnormalities reported in ≥10% pts were creatinine clearance decrease (39%), hemoglobin decrease (38%), Lymphocytes decrease (37%), ALT increase (14%), AST increase (13%), bilirubin increase (12%), and creatinine increase (12%). AE of gr 1 sinus bradycardia (asymptomatic) was reported in 35% and gr 2 (symptomatic, no intervention needed) was reported in 5%. Gr 2 and 3 QTcF prolongation were reported in 2 and 3 pts, respectively. There were no treatment-related deaths. Efficacy estimates are presented in Table 1. CI: Confidence interval. N: total number of pts in full analysis set, used in PFS analysis. n: number of response-evaluable pts, used in ORR and CBR analysis. Efficacy estimates were consistent across the various subgroups. Responses were demonstrated in heavily pretreated pts, pts with visceral metastases, pts with constitutionally active ESR1 Y537S mutations, and in pts who received chemotherapy in the metastatic setting. Numerically higher response rate and longer PFS were observed in pts with ESR1 Y537s. Conclusions: H3B-6545 has a manageable safety profile and demonstrated single-agent anti-tumor activity in heavily pretreated ER+, HER2- mBC patients. Clinical activity was consistent across the various subgroups. Tumors harboring the constitutionally active ESR1 Y537S mutation may present higher ERα activity, and consequently enrich for luminal A traits and demonstrate greater lineage dependence on ERα. Table 1.Consistency of H3B-6545 activity across the key subgroupsEfficacyClinical CharacteristicORR % (95% CI)CBR % (95% CI)Median PFS mo (95% CI)Overall population (N=94, n=72)16.7 (8.9, 27.3)40.3 (28.9, 52.5)5.1 (3.2, 6.2)Liver and/or lung metastases (N=76, n=62)17.7 (9.2, 29.5)41.9 (29.5, 55.2)5.4 (1.8, 7.2)≥3 prior regimens (N=49, n=39)20.5 (9.3, 36.5)48.7 (32.4, 65.2)5.4 (3.5, 7.3)Prior chemotherapy (N=47, n=35)17.1 (6.6, 33.6)51.4 (34.0, 68.6)5.5 (3.6, 7.3)PgR+ (N=38, n=32)15.6 (5.3, 32.8)50.0 (31.9, 68.1)5.4 (2.0, 8.8)ESR1 clonal Y537S (N=10, n=10)30.0 (6.7, 65.2)60.0 (26.2, 87.8)7.3 (0.8, 11.2)ESR1 clonal D538G (N=19, n=17)0.0 (0.0, 19.5)35.3 (14.2, 61.7)5.4 (1.7, 7.2) Citation Format: Erika P Hamilton, Judy S Wang, Timothy Pluard, Aki Morikawa, E Claire Dees, Robert H Jones, Barbara Haley, Anne Armstrong, Adam L Cohen, Pamela Munster, Gail S Wright, Fadi Kayali, Lisa Cantagallo, Manav Korpal, Jenny Long, Jianjun Xiao, Benoit Destenaves, Lei Gao, Tarek Sahmoud, Antonio Gualberto, Dejan Juric. H3B-6545, a novel selective estrogen receptor covalent antagonist (SERCA), in estrogen receptor positive (ER+), human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer - A phase II study [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P1-17-10.
- Published
- 2022
14. Abstract P5-13-18: Upregulation of immune response biomarkers by ribociclib plus endocrine therapy (ET) in paired tumor samples from phase I studies
- Author
-
Dejan Juric, Chong Ma, Ralph Tiedt, Yoon-Sim Yap, Joanne Chiu, Pamela Munster, Roohi Ismail-Khan, Laura Garcia-Estevez, Ingrid A. Mayer, Carlos Becerra, Nadia Solovieff, Agnes Lteif, Faye Su, and Yen-Shen Lu
- Subjects
Cancer Research ,Oncology - Abstract
Background: In addition to inhibiting cell cycle progression, CDK4/6 inhibitors have been found to modulate anti-cancer immune response (Goel S, et al. Nature. 2017;548:471-475). Data from preclinical studies suggested multiple mechanisms—involving changes in both cancer cells and immune cells—by which CDK4/6 inhibition can enhance anti-cancer immunity. These mechanisms include increased antigen presentation, interferon response triggering, and regulatory T-cell suppression. Clinical correlates to preclinical observations have so far been obtained using paired biopsies from neoadjuvant breast cancer trials (Johnston S, et al. J Clin Oncol. 2019;37:178-189; Hurvitz S, et al. Clin Cancer Res. 2020; ;26:566-80). Here, we describe a gene expression analysis of paired pre- and on-treatment biopsies from CLEE011A2115C and CLEE011X2107, two phase I clinical trials evaluating ribociclib in combination with ET in the metastatic breast cancer (MBC) setting. Methods: The nCounter PanCancer IO 360 Panel (NanoString) was used to quantify expression of 770 genes in 7 pairs of tumor samples (baseline vs. cycle 1 day 15) from two phase I trials in the MBC setting (5 patients from CLEE011A2115C and 2 from CLEE011X2107). All patients were treated with ribociclib and an ET. Pairwise differential gene expression analysis of individual genes and previously published immune-related gene signatures was conducted (Bedognetti D, et al. Curr Opin Oncol. 2015;27:433-444). Results: Many genes that were markedly suppressed with treatment (eg, MKI67, MYC, CDK2, CCNB1, TYMS, and DNMT1) were related to proliferation, DNA replication, and G1/S transition of the cell cycle, as expected. Interestingly, expression of numerous genes involved in immune response was increased. These were mostly interferon-regulated genes; particularly, a gene signature indicative of a T cell-inflamed tumor microenvironment (Ayers M, et al. J Clin Invest. 2017;127:2930-2940) was upregulated. Immune-related genes tended to preferentially increase with treatment in patients who later experienced a clinical response. Since bulk RNA from tumor biopsies was analyzed, we could not distinguish whether the observed expression changes reflected increased infiltration by immune cells or an endogenous interferon response in cancer cells. The latter could be triggered by DNMT1 suppression (as previously described), which then promotes T cell-mediated immunity by increasing antigen presentation and chemokine production in cancer cells. Conclusions: This analysis was conducted with a small sample size and is considered hypothesis generating; further investigation is needed. Cell cycle and proliferation markers showed robust suppression by ribociclib-based treatment regimens, as expected. Conversely, a notable increase in immune-related genes was detected that tended to occur preferentially in patients who later experienced more favorable clinical outcomes. Both changes occurred early (cycle 1 day 15). To our knowledge, this is the first clinical biomarker report of immune activation mediated by CDK4/6 inhibition in MBC. Citation Format: Dejan Juric, Chong Ma, Ralph Tiedt, Yoon-Sim Yap, Joanne Chiu, Pamela Munster, Roohi Ismail-Khan, Laura Garcia-Estevez, Ingrid A. Mayer, Carlos Becerra, Nadia Solovieff, Agnes Lteif, Faye Su, Yen-Shen Lu. Upregulation of immune response biomarkers by ribociclib plus endocrine therapy (ET) in paired tumor samples from phase I studies [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 P5-13-18.
- Published
- 2022
15. Abstract CT222: Multimodal mechanism of action of the GSK-3 inhibitor 9-ING-41 (elraglusib) includes an immunomodulatory component: preliminary results from the 1801 phase 1/2 trial
- Author
-
Taylor Weiskittel, Joseph McDermott, Daniel Billadeau, Hu Li, Anwaar Saeed, Devalingam Mahalingam, Solmaz Sahebjam, Pamela Munster, Ludimila Cavalcante, Frank J. Giles, Andrew Mazar, and Benedito Carneiro
- Subjects
Cancer Research ,Oncology - Abstract
9-ING-41 (elraglusib) is a potent and selective GSK-3 inhibitor that has shown anti-tumor activity in patient-derived xenograft models and phase1/2 clinical studies in patients (pts) with advanced solid tumors. Preclinical studies have demonstrated that elraglusib downregulates PD-1, TIGIT and LAG-3, upregulates expression of MHC class I proteins in tumor cells from “cold tumors”, and shows synergy when combined with PD-1 blockade in mouse xenografts. Clinical activity has been observed both as a single agent and in combination with standard of care chemotherapies in several advanced cancer histologies. Here we present initial ‘omics data from our phase 1/2 study (NCT03678883; ACT1801) spanning >100 pts with advanced cancer evaluating both single agent and chemotherapy combinations. The patients (pts) included in this study received elraglusib as a ≥3rd line therapy for advanced disease. Most pts with melanoma treated with elraglusib monotherapy (8/9; 89%) stayed on study for >2 cycles. One patient with melanoma refractory to checkpoint inhibitors achieved a confirmed and durable CR. Clinical benefit was also observed among pts with colorectal cancer with 4/12 (33.3%) pts treated with monotherapy and 12/15 (80%) that received elraglusib plus irinotecan rechallenge stayed on study >2 cycles reaching median overall survival of 106 and 211 days, respectively. Based on emerging in vivo and in vitro results demonstrating that elraglusib activates T and NK cells promoting anti-tumor immune responses, we hypothesize immunomodulation by elraglusib may be contributing to anti-tumor immune response in the 1801 trial. We have acquired TCRseq and RNAseq profiles of PBMC samples from seven patients in 1801 during the first two weeks of treatment with elraglusib monotherapy. In these pts, reduced TCR clonality was observed and specific TCR clonotypes expanded after treatment, indicating T cell activation and expansion. These pts also showed changes in PBMC populations during elraglusib therapy as measured by immune deconvolution of PBMC RNAseq. Taken together, these data support a novel, previously unrecognized immunomodulatory mechanism of action for elraglusib and could provide rationale for future clinical development of elraglusib in pts with advanced malignancies. Additional analysis from TCRseq, immune profiling and cytokine analysis from expanded cohort of pts is ongoing. Citation Format: Taylor Weiskittel, Joseph McDermott, Daniel Billadeau, Hu Li, Anwaar Saeed, Devalingam Mahalingam, Solmaz Sahebjam, Pamela Munster, Ludimila Cavalcante, Frank J. Giles, Andrew Mazar, Benedito Carneiro. Multimodal mechanism of action of the GSK-3 inhibitor 9-ING-41 (elraglusib) includes an immunomodulatory component: preliminary results from the 1801 phase 1/2 trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT222.
- Published
- 2023
16. Abstract GS3-03: GS3-03 ARV-471, a PROTAC® estrogen receptor (ER) degrader in advanced ER-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer: phase 2 expansion (VERITAC) of a phase 1/2 study
- Author
-
Anne F. Schott, Sara Hurvitz, Cynthia Ma, Erika Hamilton, Rita Nanda, George Zahrah, Natasha Hunter, Antoinette R. Tan, Melinda Telli, Jesus Anampa Mesias, Rinath Jeselsohn, Pamela Munster, Haolan Lu, Richard Gedrich, Cecile Mather, Janaki Parameswaran, and Hyo S. Han
- Subjects
Cancer Research ,Oncology - Abstract
Background: ARV-471 is a selective, orally administered PROteolysis TArgeting Chimera (PROTAC®) protein degrader that targets wild-type and mutant ER. ARV-471 is being evaluated in patients with ER+/HER2- locally advanced or metastatic breast cancer in a first-in-human phase 1/2 study (NCT04072952). In the phase 1 dose escalation, ARV-471 monotherapy (dose range: 30–700 mg total daily dose) showed a manageable safety profile in patients who had previously received endocrine therapy and a cyclin-dependent kinase (CDK) 4/6 inhibitor. The clinical benefit rate (CBR; rate of confirmed complete or partial response or stable disease ≥24 weeks) was 40% (95% CI: 26–56) in 47 evaluable patients. The phase 2 expansion portion of the study (VERITAC) evaluated 2 doses of ARV-471.Methods: In VERITAC, ARV-471 monotherapy was administered at doses of 200 mg once daily (QD) or 500 mg QD to patients with ER+/HER2- locally advanced/metastatic breast cancer who had received ≥1 prior endocrine therapy for ≥6 months, ≥1 CDK4/6 inhibitor, and ≤1 chemotherapy regimen. The primary endpoint of CBR was evaluated in patients enrolled ≥24 weeks prior to the data cutoff. Results: As of June 6, 2022, 71 patients received ARV-471 (200 mg QD [n=35]; 500 mg QD [n=36]) in VERITAC. Across all treated patients, 69 (97.2%) were female and median age was 60 y (range: 41–86). Patients had received a median of 4 prior regimens in all settings (range: 1–10); 100% had prior CDK4/6 inhibitors, 78.9% had prior fulvestrant, and 73.2% had prior chemotherapy. ARV-471 was well tolerated at both doses, with most treatment-related adverse events (TRAEs) grade 1/2; the most common TRAEs were fatigue and nausea (Table). In all, 3 patients (1 in the 200 mg QD cohort and 2 in the 500 mg QD cohort) discontinued ARV-471 due to treatment-emergent adverse events (TEAEs); 3 patients had ARV-471 dose reductions due to TEAEs (all from 500 mg QD to 400 mg QD). CBR was 37.1% (95% CI: 21–55) in 35 evaluable patients treated at 200 mg QD and 38.9% (95% CI: 23–57) in 36 evaluable patients treated at 500 mg QD. CBR in evaluable patients with mutant ESR1 in the 200 mg QD (n=19) and 500 mg QD (n=22) cohorts was 47.4% (95% CI: 24–71) and 54.5% (95% CI: 32–76), respectively. Conclusions: In the phase 2 VERITAC expansion cohorts of patients with ER+/HER2- locally advanced/metastatic breast cancer and prior CDK4/6 inhibitor treatment, ARV-471 monotherapy showed evidence of clinical activity based on CBR, which was further enhanced in the subgroup with ESR1 mutations. The manageable AE profile observed in the phase 1 portion of the study was maintained during cohort expansion at doses of 200 mg QD and 500 mg QD. Additional analyses are ongoing.Table. TRAEs reported in ≥10% of patients overall aNo grade 3/4 TRAE occurred in >1 patient. AST=aspartate aminotransferase Citation Format: Anne F. Schott, Sara Hurvitz, Cynthia Ma, Erika Hamilton, Rita Nanda, George Zahrah, Natasha Hunter, Antoinette R. Tan, Melinda Telli, Jesus Anampa Mesias, Rinath Jeselsohn, Pamela Munster, Haolan Lu, Richard Gedrich, Cecile Mather, Janaki Parameswaran, Hyo S. Han. GS3-03 ARV-471, a PROTAC® estrogen receptor (ER) degrader in advanced ER-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer: phase 2 expansion (VERITAC) of a phase 1/2 study [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 GS3-03.
- Published
- 2023
17. PD54-11 RESULTS OF THE PHASE 1 SAFETY AND EFFICACY PROSTATE SPECIFIC MEMBRANE ANTIGEN (PSMA) TARGETING FLUOROPHORE FOR IMAGE GUIDED SURGERY IN PATIENTS UNDERGOING ROBOTIC PROSTATECTOMY
- Author
-
Hao Nguyen, Alexander Antaris, Nynke Van den Berg, Lingru Xue, Scott Greenberg, Anna Muchnik, Alwin Klaassen, Jeff Simko, Pamela Munster, Sanjeev Dutta, Jonathan Sorger, and Peter Carroll
- Subjects
Urology - Published
- 2022
18. Prolonged Disease Stabilization and Tolerability in a Nuclear Protein in Testis Midline Carcinoma Patient Treated with Dual Histone Deacetylase and Phosphoinositide 3-Kinase Inhibitor CUDC-907
- Author
-
Meaghan McMahon, David Tuck, Pamela Munster, Robert Gharavi, and Nilson Wu
- Subjects
0301 basic medicine ,Pleural effusion ,business.industry ,Phosphoinositide 3-kinase inhibitor ,General Medicine ,medicine.disease ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,030220 oncology & carcinogenesis ,Carcinoma ,medicine ,Cancer research ,Histone deacetylase ,Nuclear protein ,business ,PI3K/AKT/mTOR pathway - Abstract
Introduction: Nuclear protein in testis midline carcinoma (NMC) is a rare and extremely aggressive carcinoma (median survival < 7 months) with no effective treatment options. CUDC-907 is a novel small molecule inhibitor of histone deacetylase (HDAC) and phosphoinositide 3-kinase (PI3K) enzymes currently being investigated in multiple tumor types, including NMC. Case Report: A 61-year old female NMC patient enrolled on study CUDC-907-102 (NCT02307240) after rapidly progressing through two prior treatments. The patient’s assessable sites of disease consisted of right pleural effusion, right hilar soft tissue, and segment IV liver. Treatment was well tolerated with toxicities primarily consisting of manageable diarrhea and thrombocytopenia. The patient remains on active treatment after more than 32 months of stable disease. Discussion: Dysregulation of MYC in NMC is believed to play a central role in pathogenesis. CUDC-907 has demonstrated potent suppression of MYC expression and anti-tumor activity in preclinical NMC models, providing a mechanistic rationale for the prolonged disease stabilization observed here. The treatment of additional NMC patients with CUDC-907 is needed to further evaluate this promising report. Conclusion: This case demonstrates a rare success in the treatment of a devastating disease using only a novel small molecule, warranting further investigation of CUDC-907 in NMC.
- Published
- 2018
19. Abstract PD8-06: Phase I/II trial of H3B-6545, a novel selective estrogen receptor covalent antagonist (SERCA), in estrogen receptor positive (ER+), human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer
- Author
-
Erika P Hamilton, Judy S Wang, Timothy Pluard, Stephen Johnston, Aki Aki Morikawa, Claire E Dees, Robert H Jones, Barbara Haley, Anne Armstrong, Adam L Cohen, Pamela Munster, Gail Wright, Fadi Kayali, Manav Korpal, Lihua Yu, Lisa Cantagallo, Benoit Destenaves, Zhaojie Zhang, Lei Gao, Marc J Pipas, Tarek Sahmouud, Antonio Gualberto, and Dejan Juric
- Subjects
Cancer Research ,Oncology - Abstract
Background: Addition of CDK 4/6 inhibitors to endocrine therapy has provided significant improvements in outcomes for patients (pts) with metastatic breast cancer (mBC). However, acquired resistance to front-line therapy remains a challenge, and response to late lines of therapy is poor. H3B-6545 is a selective, orally available, small molecule covalent antagonist of the estrogen receptor (ERα) H3B-6545 binds covalently to a cysteine residue at position 530 of both wild-type and the constitutively active mutant ERα proteins, including Y537S. H3B-6545 demonstrated significant antitumor activity in multiple PDX breast cancer models, including those with mutated ESR1 (the gene encoding ERα).Methods: This is a multicenter phase I/II trial. The primary objective of the phase I is to determine the maximum tolerated dose and recommended phase 2 dose (RP2D) in pretreated pts with ER+, HER2- mBC. Secondary objectives include safety and antitumor activity. The primary objective of the phase II is to estimate the objective response rate (ORR) and secondary objectives include clinical benefit rate (CBR), progression-free survival (PFS) and safety. The trial was designed to exclude a lower limit of ORR of 5% at one-sided level of significance of 0.05 and a power of 90%. Results: Between August 2017 and February 2020, 130 pts were enrolled; 47 in the Phase I part and 83 in the Phase II part of the trial. A total of 105 pts, including 73 patients on 450 mg, were response-evaluable. The phase I evaluated once daily doses from 100 to 600 mg. No dose-limiting toxicities (DLT) were observed at doses up to 450 mg and 2 DLTs were observed in 2 (grade 3 fatigue and grade 3 drug eruption) of 7 pts on the 600 mg cohort. Consequently, the dose of 450 mg was selected as the RP2D. Median age was 62 years (range: 31 to 87 years), 82% had liver and/or lung metastases, and the median number of prior therapies for metastatic disease was 3 (range: 1 to 10), with 41% of the pts receiving ≥ 4 prior therapies in the metastatic setting. Prior CDK4/6 inhibitors, fulvestrant, and chemotherapy were received by 87%, 71%, and 54% of the pts, respectively. 75 pts (58%) had detectable ESR1 mutations. As of May 31, 2020, grade 2 or higher adverse events reported in ≥10% were anemia (20%), fatigue (16%), nausea (14%), diarrhea (11%) and AST increase (11%). Three cases of grade 4 AE were reported (serum bilirubin, urinary tract obstruction, and hyponatremia), all considered related to disease progression. Grade 1 sinus bradycardia (asymptomatic) was reported in 35% and grade 2 (symptomatic, no intervention needed) was reported in 4%. Grade 2 and 3. QTcF prolongation were reported in 2 and 3 patients, respectively. There were no treatment-related deaths. In the response-evaluable group, 13 confirmed partial responses (PR, 12%. 90% confidence limits: 7.5%-19%), including 11 PRs (15%, 90% confidence limits: 8.7%-23.7) on 450 mg dose, were observed, thus achieving the primary objective of the trial. Stable disease (SD) and clinical benefit rates (≥23 weeks) were 45% and 33% respectively at 450 mg and 46% and 34%, respectively on all doses. Responses were observed in heavily pretreated pts, pts with visceral metastases and in pts who received prior fulvestrant, prior CDK4/6 inhibitor, and/or prior chemotherapy, in the metastatic setting. Three PRs (25%) and 4 SDs were observed in 12 pts in whom clonal ESR1 Y537S was the main ERα driver. Median PFS, in all pts and in pts with clonal ESR1 Y537S was 3.7 months and 7.3 months, respectively. Conclusions: H3B-6545 has a manageable safety profile and demonstrated single-agent anti-tumor activity in heavily pretreated ER+, HER2- mBC patients including those with a constitutively active clonal ESR1 Y537S mutation. Citation Format: Erika P Hamilton, Judy S Wang, Timothy Pluard, Stephen Johnston, Aki Aki Morikawa, Claire E Dees, Robert H Jones, Barbara Haley, Anne Armstrong, Adam L Cohen, Pamela Munster, Gail Wright, Fadi Kayali, Manav Korpal, Lihua Yu, Lisa Cantagallo, Benoit Destenaves, Zhaojie Zhang, Lei Gao, Marc J Pipas, Tarek Sahmouud, Antonio Gualberto, Dejan Juric. Phase I/II trial of H3B-6545, a novel selective estrogen receptor covalent antagonist (SERCA), in estrogen receptor positive (ER+), human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD8-06.
- Published
- 2021
20. TRASTUZUMAB INDUCED CARDIOTOXICITY: WITHOUT PRIOR ANTHRACYCLINES, DECREASE IN LEFT VENTRICULAR EJECTION FRACTION IS NEGLIGIBLE
- Author
-
Roy N. Tamura, Jeffrey Krischer, Maya Guglin, and Pamela Munster
- Subjects
medicine.medical_specialty ,Cardiotoxicity ,Ejection fraction ,Anthracycline ,business.industry ,Lisinopril ,Placebo ,medicine.disease ,Breast cancer ,Trastuzumab ,Internal medicine ,medicine ,Cardiology ,skin and connective tissue diseases ,Cardiology and Cardiovascular Medicine ,business ,Carvedilol ,medicine.drug - Abstract
In a randomized, double-blind, placebo-controlled trial, we enrolled HER-2 breast cancer patients into two arms depending on the anthracycline exposure prior to trastuzumab, and randomized them into placebo, lisinopril, and carvedilol. We reported the outcomes on prevention of cardiotoxicity.
- Published
- 2020
21. Coil combination methods for multi-channel hyperpolarized
- Author
-
Zihan, Zhu, Xucheng, Zhu, Michael A, Ohliger, Shuyu, Tang, Peng, Cao, Lucas, Carvajal, Adam W, Autry, Yan, Li, John, Kurhanewicz, Susan, Chang, Rahul, Aggarwal, Pamela, Munster, Duan, Xu, Peder E Z, Larson, Daniel B, Vigneron, and Jeremy W, Gordon
- Subjects
Carbon Isotopes ,Phantoms, Imaging ,Reproducibility of Results ,Breast Neoplasms ,Signal-To-Noise Ratio ,Magnetic Resonance Imaging ,Article ,Molecular Imaging ,Electromagnetic Fields ,Neoplasms ,Pyruvic Acid ,Image Processing, Computer-Assisted ,Humans ,Computer Simulation ,Female ,Algorithms - Abstract
Effective coil combination methods for human hyperpolarized (13)C spectroscopy multi-channel data had been relatively unexplored. This study implemented and tested several coil combination methods, including 1) the sum-of-squares (SOS), 2) singular value decomposition (SVD), 3) Roemer method by using reference peak area as a sensitivity map (RefPeak), and 4) Roemer method by using ESPIRiT-derived sensitivity map (ESPIRiT). These methods were evaluated by numerical simulation, thermal phantom experiments, and human cancer patient studies. Overall, the SVD, RefPeak, and ESPIRiT methods demonstrated better accuracy and robustness than the SOS method. Extracting complex pyruvate signal provides an easy and excellent approximation of the coil sensitivity map while maintaining valuable phase information of the coil-combined data.
- Published
- 2018
22. Twisting Fate : My Journey with BRCA—from Breast Cancer Doctor to Patient and Back
- Author
-
Pamela Munster and Pamela Munster
- Subjects
- Breast--Cancer--Patients--Biography, Women physicians--Biography, Breast--Cancer--Treatment
- Abstract
From a woman who's made her living researching breast cancer—and who lived through it herself—a personal yet practical guide to the medical and emotional facets of this life-changing diagnosis A leading oncologist at the University of California San Francisco, Dr. Pamela Munster has advised thousands of women on how to cope with the realities of breast cancer, from diagnosis through treatment and recovery. But her world turned upside down when, at forty-eight years old and in otherwise perfect health, she got a call saying that her own mammogram showed “irregularities.” That single word thrust her into a wholly new role—as patient, and not only that of cancer but of the feared BRCA gene mutation as well. Suddenly, she realized that being a true “expert” in a disease was far beyond the scope of her medical training, and that she had a lot to learn if she wanted to hold onto her precious life. Weaving together her personal story with groundbreaking research on BRCA—responsible for breast cancer and many other inherited cancers affecting both women and men—Twisting Fate is an inspiring guide to living with the uncertainties of cancer. With authority, insight, and compassion, Dr. Munster uses her voice to create a safe space for genuine healing and honesty in a world otherwise too-often dominated by fear—and she is living proof of how important it is to embrace all the twists and turns of fate.
- Published
- 2018
23. CHEMOTHERAPY-INDUCED CARDIOMYOPATHY: ANTHRACYCLINES AND TRASTUZUMAB REQUIRE DIFFERENT APPROACH TO MONITORING AND PREVENTION
- Author
-
Pamela Munster, Jeffrey Krischer, Maya Guglin, Angelina Fink, Lauren Bello-Matricaria, and Roy N. Tamura
- Subjects
Drug ,Chemotherapy induced cardiomyopathy ,medicine.medical_specialty ,Ejection fraction ,biology ,business.industry ,media_common.quotation_subject ,Angiotensin-converting enzyme ,medicine.disease ,humanities ,Breast cancer ,Trastuzumab ,Internal medicine ,cardiovascular system ,Cardiology ,biology.protein ,Medicine ,cardiovascular diseases ,skin and connective tissue diseases ,Cardiology and Cardiovascular Medicine ,business ,Beta (finance) ,media_common ,medicine.drug - Abstract
Trastuzumab is a highly effective drug for breast cancer, but it is cardiotoxic, especially if used after anthracyclines. Monitoring of left ventricular ejection fraction (LVEF) is required. Several studies found benefit of angiotensin converting enzyme inhibitors and beta blockers for prevention of
- Published
- 2019
24. Interest in initiating an early phase clinical trial: results of a longitudinal study of advanced cancer patients
- Author
-
Laura B, Dunn, Jim, Wiley, Sarah, Garrett, Fay, Hlubocky, Christopher, Daugherty, Laura, Trupin, Pamela, Munster, and Daniel, Dohan
- Subjects
Adult ,Male ,Clinical Trials as Topic ,Physician-Patient Relations ,Decision Making ,Patient Preference ,Middle Aged ,Neoplasms ,Quality of Life ,Humans ,Female ,Longitudinal Studies ,Patient Participation ,Aged - Abstract
Enhanced recruitment of patients with advanced cancer (ACP) to early phase (EP) trials is needed. However, selective recruitment may affect the kinds of patients who are recruited. To address whether ACP who initiate EP trial enrollment differ from those who do not, we prospectively surveyed ACP well in advance of potential trial recruitment and followed them over time to identify those who initiated the recruitment process.EP trial initiation was defined as a patient being referred for screening to an active EP trial. Depression and anxiety were assessed with the Patient Health Questionnaire (PHQ-9) and Generalized Anxiety Disorder Scale (GAD-7), respectively. Demographic and disease characteristics, functional status, and patient preferences regarding decision making were examined as possible predictors of EP trial initiation.Of the 78 advanced cancer patients in the cohort studied, 21 (27%) initiated EP trial participation, while 57 (73%) did not. Of those who initiated this process, 14 (67%) went on to enroll in an EP study. Level of depression severity was associated with EP trial initiation, with rates of initiation nearly three times higher (35% vs. 12%, p = 0.054) among patients with minimal to mild levels of depression compared to those with moderate or higher levels of depression. EP trial initiation was not associated with demographic or socioeconomic variables, cancer type, functional status, quality of life, or decision-making variables.The presence of elevated depressive symptoms may be associated with the EP trial recruitment and enrollment processes. This possible relationship warrants further study. Copyright © 2016 John WileySons, Ltd.
- Published
- 2015
25. Contributors
- Author
-
Masahiko Abematsu, David W. Anderson, Roberta H. Andronikos, Adriana Arita, Zoya Avramova, Autumn J. Bernal, Javier Campión, Vince Castranova, Frances A. Champagne, Wendy Chao, Fei Chen, Xiaowei Sylvia Chen, Xiaodong Cheng, Lesley Collins, Max Costa, James P. Curley, Walter Doerfler, Tamar Dvash, Thomas Eggermann, Guoping Fan, Tamara B. Franklin, Steffen Gay, Eric Gilson, Johannes Gräff, Hideharu Hashimoto, Naoko Hattori, Zdenko Herceg, Norbert Hochstein, John R. Horton, Simon H. House, Karolina Janitz, Michal Janitz, Randy L. Jirtle, Ji-Hoon E. Joo, Berry Juliandi, Astrid Jungel, Hong Kan, Tae Hoon Kim, Yoon Jung Kim, Hervé Lalucque, Sheila C.S. Lima, Sally A. Litherland, John C. Lucchesi, Hanna Maciejewska-Rodrigues, Frédérique Magdinier, Fabienne Malagnac, Isabelle M. Mansuy, J. Alfredo Martinez, Rahia Mashoodh, Fermin I. Milagro, Ashleigh Miller, Pamela Munster, Susan K. Murphy, Rabih Murr, Kinichi Nakashima, Anja Naumann, Alexandre Ottaviani, Jacob Peedicayil, Gerd P. Pfeifer, Luis Felipe Ribeiro Pinto, Vincenzo Pirrotta, Pier Lorenzo Puri, Tibor A. Rauch, Lee B. Riley, Eric D. Roth, Tania L. Roth, Richard Saffery, Vittorio Sartorelli, Caroline Schluth-Bolard, Barbara Schönfeld, Axel Schumacher, Philippe Silar, Kjetil Søreide, J. David Sweatt, Scott Thomas, Kenneth T. Thurn, Trygve O. Tollefsbol, Toshikazu Ushijima, David A. Wacker, Stefanie Weber, and Xing Zhang
- Published
- 2011
26. Cognitive functioning after adjuvant chemotherapy and/or radiotherapy for early-stage breast carcinoma
- Author
-
Michael A. Andrykowski, Paul B. Jacobsen, Brent J. Small, Frederick A. Schmitt, Kristine A. Donovan, and Pamela Munster
- Subjects
Cancer Research ,Pediatrics ,medicine.medical_specialty ,medicine.medical_treatment ,Breast Neoplasms ,Motor Activity ,Article ,Cognition ,Quality of life ,Memory ,Surveys and Questionnaires ,medicine ,Prevalence ,Humans ,Attention ,Cognitive skill ,Neoadjuvant therapy ,Language ,Neoplasm Staging ,Gynecology ,Analysis of Variance ,business.industry ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Neoadjuvant Therapy ,Radiation therapy ,Oncology ,Chemotherapy, Adjuvant ,Quality of Life ,Early-Stage Breast Carcinoma ,Female ,Breast carcinoma ,business ,Cognition Disorders ,Chemo brain - Abstract
Each year in the U.S. approximately 200,000 women are diagnosed with breast carcinoma.1 The majority are diagnosed in the early stages and 90% of women can expect to live 5 years or more.1 This high rate of survivorship has promoted an ever-increasing interest in quality of life. In particular, systematic efforts have been made to identify and then minimize longer-term side effects of treatments. Recently, there has been growing recognition that women diagnosed with early-stage breast carcinoma may experience cognitive problems as a consequence of adjuvant chemotherapy.2,3 To our knowledge, eight studies to date 4–11 have formally investigated the relationship between cognitive functioning and the receipt of chemotherapy in women treated for early-stage breast carcinoma. In general, findings from these studies suggest women who receive adjuvant chemotherapy are likely to experience some degree of cognitive dysfunction. These women also tend to report more cognitive problems. However, methodological limitations of existing research make it difficult to conclude that these difficulties are primarily a consequence of chemotherapy administration. In some studies, data from breast carcinoma patients have been compared with published norms4 or healthy females with no history of breast carcinoma.8 These comparisons fail to account for the possibility that the difficulties observed are due to the diagnosis of breast carcinoma and are not specific to chemotherapy administration. In those studies that have compared cognitive functioning in breast carcinoma patients treated with or without chemotherapy, the results are mixed. Three studies found breast carcinoma patients treated with chemotherapy performed poorer than breast carcinoma patients not treated with chemotherapy,5,6,9 whereas two studies did not.7,10 Limiting conclusions further, three of these studies5–7 included the same group of women; that is, the group of breast carcinoma patients not treated with chemotherapy were identical across the three studies published by the same research group. Most existing research includes small sample sizes,4 –9,11 which limits the statistical power to determine true differences. With only one exception,11 women with breast carcinoma have been recruited to participate in these studies only after completing treatment. Whether women who agree to participate in a study of cognitive function are more likely to demonstrate cognitive deficits and to report more cognitive complaints is not known. Finally, the timing of cognitive assessments has been far from consistent across studies, ranging from 2 weeks postchemotherapy4 to 10 years after diagnosis.9 Even within studies, participants have been assessed across a wide range of time elapsed since treatment completion.4 – 8 To address these limitations, we recruited a sample of women newly diagnosed with early-stage breast carcinoma before they began adjuvant treatment. Women were evaluated approximately 6 months after completing treatment. As in most previous studies,4 –7,9,11 we used a standard battery of neuropsychologic measures to assess several domains of cognitive functioning. Our primary aim was to determine whether there were differences in cognitive functioning between women treated with and without chemotherapy. Based on previous research, we hypothesized that women who received chemotherapy would demonstrate poorer cognitive functioning than women who did not.
- Published
- 2005
27. Abstract A120: A Phase Ib study of CC-486 (Oral Azacitidine) as a priming agent for carboplatin or NAB-paclitaxel in subjects with relapsed and refractory solid tumors
- Author
-
Patricia LoRusso, Drew Rasco, Johanna Bendell, Jasgit Sachdev, Ramesh Ramanathan, Glenn Weiss, Pamela Munster, William J. Edenfield, Kejian Liu, Anne Blackwood-Chirchir, Jorge DiMartino, and Daniel D. Von Hoff
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Anemia ,Nausea ,Neutropenia ,medicine.disease ,Gastroenterology ,Carboplatin ,Oral Azacitidine ,chemistry.chemical_compound ,Peripheral neuropathy ,Oncology ,chemistry ,Internal medicine ,Immunology ,medicine ,Vomiting ,medicine.symptom ,business ,Adverse effect - Abstract
Background: CC-486, an oral formulation of 5-azacitidine, induces global genomic hypomethylation and clinical responses in MDS patients at doses of 200 mg and 300 mg/day for 14 or 21/28 days with acceptable safety. This non-randomized, multi-center Phase Ib study was designed to determine whether CC-486 can be delivered safely as priming for carboplatin or NAB-Paclitaxel (ABI-007) in patients with solid tumors. Methods: In Part 1 of this study, patients with refractory solid tumors were assigned to Arm A: CC-486 days 1 - 14 with carboplatin AUC 4 on day 8 of a 21 day cycle (n = 13) or Arm B: CC-486 days 1 - 14 with ABI-007 100 mg/m2 given weekly starting on Day 8 of Cycle 1 (n = 24). 200 mg and 300 mg dose levels (DLs) of CC-486 were evaluated in cohorts of 6 to identify a recommended Part 2 dose (RP2D). Results: On Arm A, 200 mg and 300 mg doses of CC-486 were tolerated. There was one DLT (Grade 3 pericardial effusion) at the 300 mg DL. Other related treatment-emergent adverse events (TEAEs) included anemia (53.8%) and neutropenia (46.3%). On Arm B, there were 2 DLTs of neutropenia at the 200 mg DL. The protocol was amended to give ABI-007 on days 8 and 15/21. No DLTs were encountered on 200 mg of CC-486 with intermittent ABI-007. At 300 mg, the MTD was exceeded with 2 DLTs of neutropenia (cholangiocarcinoma, pancreatic). Other common TEAEs in Arm B included nausea/vomiting (45.8%) and peripheral neuropathy (50%). Evidence of activity seen at both DLs included hypomethylation in PBMCs, 3 PRs in Arm B and 5 subjects with stable disease (> 4 mos) in Arm A. CC-486 plasma exposure showed high interpatient variability but was higher at 300 mg than 200 mg. Incidence and severity of AEs did not differ significantly between the 2 DLs. 300 mg was selected as the RP2D in combination with carboplatin and 200 mg in combination with ABI-007. Conclusion: CC-486 dosed 14/21 days is tolerated as a priming agent with carboplatin and ABI-007. Both combinations show evidence of clinical activity. Expansion cohorts at the RP2D of Arm A (relapsed/refractory bladder or ovarian carcinoma) and Arm B (relapsed/refractory NSCLC or pancreatic carcinoma) are enrolling to further characterize safety, PK/PD and anti-tumor activity. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A120. Citation Format: Patricia LoRusso, Drew Rasco, Johanna Bendell, Jasgit Sachdev, Ramesh Ramanathan, Glenn Weiss, Pamela Munster, William J. Edenfield, Kejian Liu, Anne Blackwood-Chirchir, Jorge DiMartino, Jorge DiMartino, Daniel D. Von Hoff. A Phase Ib study of CC-486 (Oral Azacitidine) as a priming agent for carboplatin or NAB-paclitaxel in subjects with relapsed and refractory solid tumors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A120.
- Published
- 2013
28. Multicenter Phase I Trial of Intraventricular Immuno-Chemotherapy in Recurrent CNS Lymphoma
- Author
-
James L. Rubenstein, Lingjing Chen, Ranjana Advani, Jan Drappatz, Elizabeth Gerstner, Tracy Batchelor, Hendrikus Krouwer, Cigall Kadoch, Pamela Munster, Soonmee Cha, Marc A Shuman, and Lloyd E. Damon
- Subjects
immune system diseases ,hemic and lymphatic diseases ,Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Abstract 959 Background: We recently studied the safety and activity of intraventricular rituximab monotherapy in the treatment of recurrent intraocular and CNS non-Hodgkin lymphoma (NHL) (JCO 2007 25: 1350–1356). Rapid dissemination throughout the craniospinal axis was demonstrated and cytologic responses were detected in six out of ten patients. Two patients experienced improvement in intraocular NHL and one exhibited resolution of parenchymal NHL. The major goals of this current trial are to perform the first study of intraventricular immuno-chemotherapy in humans and to evaluate the safety of two dose levels of intraventricular rituximab as well as its pharmacokinetics in combination with intraventricular methotrexate (MTX). Secondary goals are to obtain information regarding the efficacy of this approach in the treatment of patients with recurrent CNS lymphoma, (brain parenchyma, intraocular or the leptomeningeal compartment). Methods: Thirteen patients with recurrent/refractory CD20+ CNS NHL were treated at UCSF, Dana Farber Cancer Institute and Massachusetts General Hospital on a phase I clinical trial involving twice weekly intraventricular rituximab at 10 mg and 25 mg dose levels, administered over a planned four week schedule. Rituximab is combined with MTX (12 mg) during the second intraventricular injection each week. Patients achieving partial response or better were eligible for a second month of combination intraventricular rituximab plus MTX. Results: No serious treatment-related toxicity has been detected with intraventricular rituximab/MTX at the 10 mg and 25 mg dose levels. The most common treatment-related toxicity was paresthesias (grade 1). One patient exhibited NHL progression outside of the CNS at three weeks and thus was not evaluable for CNS response, and one patient exhibited stable disease. Complete cytologic responses were detected in ten out of thirteen patients at the one-month restaging. Parenchymal responses were detected in three out of six subjects with one partial response within the corpus callosum, and two complete regressions of lesions within temporal lobe and cerebral cortex. One patient with leptomeningeal lymphoma non-responsive to intravenous rituximab and refractory to high-dose intravenous and intrathecal methotrexate, oral temozolomide and intrathecal liposomal cytarabine, obtained a sustained complete response with the intraventricular rituximab/MTX protocol and subsequently was approved for consolidative autologous stem cell transplant. Two patients have participated in extended dosing on protocol without progression for five and eight months respectively. Thus far, there is no evidence for a relationship between FcGR3A polymorphism and therapeutic resistance in a preliminary analysis (n=8 patients). The maximum tolerated dose of intraventricular rituximab with this combination was established as 25 mg. The mean maximum intraventricular rituximab concentration after intraventricular injection of rituximab was 285 microgram/ml at the 10 mg dose level (N=3) and 882 microgram/ml at the 25 mg dose level (N=10); (p Conclusions: Intraventricular rituximab/MTX appears to be safe in recurrent CNS NHL. Twelve of thirteen patients completed at least one month of therapy, without any treatment-associated serious adverse events at any of the institutions. Intraventricular administration of methotrexate may significantly attenuate rituximab clearance from the leptomeningeal compartment. There is encouraging evidence for significant activity of intraventricular immuno-chemotherapy in the treatment of drug-resistant CNS NHL, refractory or non-responsive to intravenous rituximab. These results provide strong support for further investigation of this novel therapeutic approach. NCT00221325. Supported by Leukemia and Lymphoma Society and NIH Grant CA13908301. Disclosures: Off Label Use: We will discuss the off-label use of rituximab within the leptomeningeal compartment to treat recurrent/refractory CNS lymphomas. Advani:Seattle Genetics: Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2011
29. Olaparib and ASTX727 in BRCA1/2- and Homologous Recombination Deficient (HRD)-Mutated Tumors
- Author
-
National Cancer Institute (NCI) and Pamela Munster, Principal Investigator
- Published
- 2024
30. Pazopanib in Combination With PCI-24781 in Patients With Metastatic Solid Tumors
- Author
-
Pharmacyclics LLC., Novartis, Xynomic Pharmaceuticals, Inc., GlaxoSmithKline Research and Education Foundation for Cardiovascular Disease, and Pamela Munster, Professor, Department of Medicine (Hematology/Oncology), UCSF
- Published
- 2024
31. Combination Therapy in Cancers With Mutations in DNA Repair Genes
- Author
-
GlaxoSmithKline and Pamela Munster, Principal Investigator
- Published
- 2023
32. Alpelisib in Combination With Carboplatin in Patients With Solid Tumors and HPV-Positive Squamous Cell Carcinoma
- Author
-
Novartis and Pamela Munster, Principal Investigator
- Published
- 2022
33. Abexinostat, Palbociclib, and Fulvestrant for the Treatment of Breast or Gynecologic Cancer
- Author
-
Pfizer, Xynomic Pharmaceuticals, Inc., and Pamela Munster, Prinicpal Investigator
- Published
- 2021
34. Rucaparib and Irinotecan in Cancers With Mutations in DNA Repair
- Author
-
Clovis Oncology, Inc. and Pamela Munster, Professor, Department of Medicine; Director, Early Phase Clinical Trials Unit
- Published
- 2021
35. Cognitive functioning after adjuvant chemotherapy and/or radiotherapy for early‐stage breast carcinoma.
- Author
-
Kristine A. Donovan, Brent J. Small, Michael A. Andrykowski, Frederick A. Schmitt, Pamela Munster, and Paul B. Jacobsen
- Published
- 2005
36. Study of BMN-673 With Carboplatin and Paclitaxel in Patients With Advanced BRCA-mutated Solid Tumor or Triple Negative Metastatic Breast Cancer
- Author
-
QuantumLeap Healthcare Collaborative and Pamela Munster, Professor in Residence
- Published
- 2020
37. Phase Ib Study of Alpelisib With Cisplatin in Patients With HPV+ Solid Tumor Malignancies
- Author
-
Pamela Munster, Professor
- Published
- 2020
38. 64-Cu Labeled Brain PET/MRI for MM-302 in Advanced HER2+ Cancers With Brain Mets
- Author
-
Pamela Munster, Professor
- Published
- 2017
39. Gonadotropin-Releasing Hormone Agonists During Chemotherapy for Preservation of Ovarian Function and Fertility in Premenopausal Patients With Early Breast Cancer: A Systematic Review and Meta-Analysis of Individual Patient-Level Data.
- Author
-
Lambertini M, Moore HCF, Leonard RCF, Loibl S, Munster P, Bruzzone M, Boni L, Unger JM, Anderson RA, Mehta K, Minton S, Poggio F, Albain KS, Adamson DJA, Gerber B, Cripps A, Bertelli G, Seiler S, Ceppi M, Partridge AH, and Del Mastro L
- Subjects
- Adult, Disease-Free Survival, Female, Humans, Premenopause, Primary Ovarian Insufficiency chemically induced, Randomized Controlled Trials as Topic, Treatment Outcome, Breast Neoplasms drug therapy, Fertility Preservation methods, Gonadotropin-Releasing Hormone agonists, Organ Sparing Treatments methods, Ovary drug effects, Primary Ovarian Insufficiency prevention & control
- Abstract
Purpose The role of temporary ovarian suppression with gonadotropin-releasing hormone agonists (GnRHa) during chemotherapy as a strategy to preserve ovarian function and fertility in premenopausal women remains controversial. This systematic review and meta-analysis using individual patient-level data was conducted to better assess the efficacy and safety of this strategy in patients with early breast cancer. Methods The trials in which premenopausal women with early breast cancer were randomly assigned to receive (neo)adjuvant chemotherapy alone or with concurrent GnRHa were eligible for inclusion. Primary end points were premature ovarian insufficiency (POI) rate and post-treatment pregnancy rate. Disease-free survival and overall survival were secondary end points. Because each study represents a cluster, statistical analyses were performed using a random effects model. Results A total of 873 patients from five trials were included. POI rate was 14.1% in the GnRHa group and 30.9% in the control group (adjusted odds ratio, 0.38; 95% CI, 0.26 to 0.57; P < .001). A total of 37 (10.3%) patients had at least one post-treatment pregnancy in the GnRHa group and 20 (5.5%) in the control group (incidence rate ratio, 1.83; 95% CI, 1.06 to 3.15; P = .030). No significant differences in disease-free survival (adjusted hazard ratio, 1.01; 95% CI, 0.72 to 1.42; P = .999) and overall survival (adjusted hazard ratio, 0.67; 95% CI, 0.42 to 1.06; P = .083) were observed between groups. Conclusion Our findings provide evidence for the efficacy and safety of temporary ovarian suppression with GnRHa during chemotherapy as an available option to reduce the likelihood of chemotherapy-induced POI and potentially improve future fertility in premenopausal patients with early breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
40. Activating NOTCH1 Mutations Define a Distinct Subgroup of Patients With Adenoid Cystic Carcinoma Who Have Poor Prognosis, Propensity to Bone and Liver Metastasis, and Potential Responsiveness to Notch1 Inhibitors.
- Author
-
Ferrarotto R, Mitani Y, Diao L, Guijarro I, Wang J, Zweidler-McKay P, Bell D, William WN Jr, Glisson BS, Wick MJ, Kapoun AM, Patnaik A, Eckhardt G, Munster P, Faoro L, Dupont J, Lee JJ, Futreal A, El-Naggar AK, and Heymach JV
- Subjects
- Adult, Aged, Animals, Bone Neoplasms drug therapy, Bone Neoplasms mortality, Bone Neoplasms secondary, Carcinoma, Adenoid Cystic drug therapy, Carcinoma, Adenoid Cystic mortality, Carcinoma, Adenoid Cystic secondary, DNA Mutational Analysis, Disease-Free Survival, Female, Genetic Predisposition to Disease, HEK293 Cells, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Liver Neoplasms drug therapy, Liver Neoplasms mortality, Liver Neoplasms secondary, Male, Mice, Nude, Middle Aged, Molecular Targeted Therapy, Phenotype, Predictive Value of Tests, Proportional Hazards Models, Receptor, Notch1 antagonists & inhibitors, Receptor, Notch1 metabolism, Retrospective Studies, Risk Factors, Salivary Gland Neoplasms drug therapy, Salivary Gland Neoplasms mortality, Salivary Gland Neoplasms pathology, Time Factors, Transfection, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Biomarkers, Tumor genetics, Bone Neoplasms genetics, Carcinoma, Adenoid Cystic genetics, Liver Neoplasms genetics, Mutation, Receptor, Notch1 genetics, Salivary Gland Neoplasms genetics
- Abstract
Purpose Adenoid cystic carcinomas (ACCs) represent a heterogeneous group of chemotherapy refractory tumors, with a subset demonstrating an aggressive phenotype. We investigated the molecular underpinnings of this phenotype and assessed the Notch1 pathway as a potential therapeutic target. Methods We genotyped 102 ACCs that had available pathologic and clinical data. Notch1 activation was assessed by immunohistochemistry for Notch1 intracellular domain. Luciferase reporter assays were used to confirm Notch1 target gene expression in vitro. The Notch1 inhibitor brontictuzumab was tested in patient-derived xenografts from patients with ACC and in a patient with ACC who was enrolled in a phase I study. Results NOTCH1 mutations occurred predominantly (14 of 15 patients) in the negative regulatory region and Pro-Glu-Ser-Thr-rich domains, the same two hotspots seen in T-cell acute lymphoblastic leukemias, and led to pathway activation in vitro. NOTCH1-mutant tumors demonstrated significantly higher levels of Notch1 pathway activation than wild-type tumors on the basis of Notch1 intracellular domain staining ( P = .004). NOTCH1 mutations define a distinct aggressive ACC subgroup with a significantly higher likelihood of solid subtype ( P < .001), advanced-stage disease at diagnosis ( P = .02), higher rate of liver and bone metastasis ( P ≤ .02), shorter relapse-free survival (median, 13 v 34 months; P = .01), and shorter overall survival (median 30 v 122 months; P = .001) when compared with NOTCH1 wild-type tumors. Significant tumor growth inhibition with brontictuzumab was observed exclusively in the ACC patient-derived xenograft model that harbored a NOTCH1 activating mutation. Furthermore, an index patient with NOTCH1-mutant ACC had a partial response to brontictuzumab. Conclusion NOTCH1 mutations define a distinct disease phenotype characterized by solid histology, liver and bone metastasis, poor prognosis, and potential responsiveness to Notch1 inhibitors. Clinical studies targeting Notch1 in a genotype-defined ACC subgroup are warranted.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.