92 results on '"Pegram MD"'
Search Results
2. Abstract P6-17-36: Consensus and disagreement among experts for treatment of patients with HER2+ early-stage breast cancer suggests unmet need for online decision support tool
- Author
-
Holmes, FA, primary, Rosenthal, KM, additional, Hurvitz, S, additional, Pegram, MD, additional, Yardley, DA, additional, Obholz, KL, additional, and O'Shaughnessy, J, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P6-05-05: Discistronic reporter screen for internal ribosome entry site (IRES) - mediated translational regulation of truncated p110 ERBB2 isoform
- Author
-
Zong, Y, primary, Li, Y, additional, Liu, X, additional, and Pegram, MD, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract OT1-02-07: SOPHIA: A phase 3, randomized study of margetuximab plus chemotherapy vs trastuzumab plus chemotherapy in the treatment of patients with HER2+ metastatic breast cancer
- Author
-
Rugo, HS, primary, Pegram, MD, additional, Gradishar, WJ, additional, Cortes, J, additional, Curigliano, G, additional, Hong, S, additional, Wigginton, JM, additional, Lechleider, RJ, additional, and Cardoso, F, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract OT3-01-04: An open-label, single-arm, phase II study of pertuzumab with high-dose trastuzumab for the treatment of central nervous system progression post-radiotherapy in patients with HER2-positive metastatic breast cancer (PATRICIA)
- Author
-
Lin, NU, primary, Pegram, MD, additional, Lai, C, additional, Lacasia, A, additional, Stein, A, additional, Yoo, B, additional, and Perez, EA, additional
- Published
- 2016
- Full Text
- View/download PDF
6. Abstract P6-04-10: Components of cap-independent translation as novel targets in breast cancer
- Author
-
Ward, TM, primary, Harrell, C, additional, Liu, X, additional, and Pegram, MD, additional
- Published
- 2013
- Full Text
- View/download PDF
7. Abstract P5-08-03: In vitro resistance to ERBB2-targeted therapies does not bestow cross-resistance to antibody dependent cellular cytotoxicity
- Author
-
Olson, RM, primary, Ward, TM, additional, Jegg, AM, additional, Liu, X, additional, and Pegram, MD, additional
- Published
- 2013
- Full Text
- View/download PDF
8. P2-01-25: Truncated p110 ERBB2 (CTF611) Increases Migration and Invasion of Breast Epithelial Cells by Inhibiting STAT5b Activation.
- Author
-
Ward, TM, primary, Iorns, E, additional, Hoe, N, additional, Kim, P, additional, Singh, S, additional, Ernani, V, additional, Liu, X, additional, Jegg, A-M, additional, Gallas, M, additional, Lippman, ME, additional, and Pegram, MD, additional
- Published
- 2011
- Full Text
- View/download PDF
9. PD01-09: Identifying Novel Mechanisms of Resistance to Lapatinib in ERBB2+ Breast Cancer Cells through Whole Genome Mutational Analysis.
- Author
-
Jegg, A, primary, Ward, TM, additional, Iorns, E, additional, Gallas, M, additional, Aparicio, SA, additional, and Pegram, MD, additional
- Published
- 2011
- Full Text
- View/download PDF
10. Phase I dose escalation and pharmacokinetic study of lapatinib in combination with trastuzumab in patients with advanced ErbB2-positive breast cancer.
- Author
-
Storniolo AM, Pegram MD, Overmoyer B, Silverman P, Peacock NW, Jones SF, Loftiss J, Arya N, Koch KM, Paul E, Pandite L, Fleming RA, Lebowitz PF, Ho PT, and Burris HA 3rd
- Published
- 2008
11. What is optimal management for breast cancer patients whose disease progresses while on trastuzumab?
- Author
-
Tuma RS, Pegram MD, and Cianfrocca ME
- Published
- 2009
- Full Text
- View/download PDF
12. Impact of the Breast Cancer Index for Extended Endocrine Decision-Making: First Results of the Prospective BCI Registry Study.
- Author
-
Sanft TB, Wong J, O'Neal B, Siuliukina N, Jankowitz RC, Pegram MD, Fox JR, Zhang Y, Treuner K, and O'Shaughnessy JA
- Subjects
- Humans, Female, Prospective Studies, Chemotherapy, Adjuvant methods, Neoplasm Recurrence, Local drug therapy, Breast Neoplasms pathology, Brain-Computer Interfaces
- Abstract
Background: The Breast Cancer Index (BCI) test assay provides an individualized risk of late distant recurrence (5-10 years) and predicts the likelihood of benefitting from extended endocrine therapy (EET) in hormone receptor-positive early-stage breast cancer. This analysis aimed to assess the impact of BCI on EET decision-making in current clinical practice., Methods: The BCI Registry study evaluates long-term outcomes, decision impact, and medication adherence in patients receiving BCI testing as part of routine clinical care. Physicians and patients completed pre-BCI and post-BCI test questionnaires to assess a range of questions, including physician decision-making and confidence regarding EET; patient preferences and concerns about the cost, side effects, drug safety, and benefit of EET; and patient satisfaction regarding treatment recommendations. Pre-BCI and post-BCI test responses were compared using McNemar's test and Wilcoxon signed rank test., Results: Pre-BCI and post-BCI questionnaires were completed for 843 physicians and 823 patients. The mean age at enrollment was 65 years, and 88.4% of patients were postmenopausal. Of the tumors, 74.7% were T1, 53.4% were grade 2, 76.0% were N0, and 13.8% were HER2-positive. Following BCI testing, physicians changed EET recommendations in 40.1% of patients (P<.0001), and 45.1% of patients changed their preferences for EET (P<.0001). In addition, 38.8% of physicians felt more confident in their recommendation (P<.0001), and 41.4% of patients felt more comfortable with their EET decision (P<.0001). Compared with baseline, significantly more patients were less concerned about the cost (20.9%; P<.0001), drug safety (25.4%; P=.0014), and benefit of EET (29.3%; P=.0002)., Conclusions: This analysis in a large patient cohort of the BCI Registry confirms and extends previous findings on the significant decision-making impact of BCI on EET. Incorporating BCI into clinical practice resulted in changes in physician recommendations, increased physician confidence, improved patient satisfaction, and reduced patient concerns regarding the cost, drug safety, and benefit of EET.
- Published
- 2024
- Full Text
- View/download PDF
13. Epidemiology, clinical outcomes, and unmet needs of patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases: A systematic literature review.
- Author
-
Müller V, Bartsch R, Lin NU, Montemurro F, Pegram MD, and Tolaney SM
- Subjects
- Humans, Female, Trastuzumab therapeutic use, Receptor, ErbB-2 metabolism, Breast Neoplasms drug therapy, Brain Neoplasms drug therapy
- Abstract
Background: There is an increasing need for developing effective therapies for managing intracranial disease in patients with human epidermal growth factor receptor 2-positive (HER2 +) metastatic breast cancer and brain metastases (BM), as this population is growing and has historically been excluded from large clinical trials. In this systematic literature review, we aimed to provide a comprehensive overview of the epidemiology, unmet needs, and global treatment landscape for patients with HER2 + metastatic breast cancer and BM, with a particular focus on heterogeneity across clinical trial designs in this setting., Methods: We conducted literature searches of PubMed and select congress websites up to March 2022 and filtered for publications with a significant focus on epidemiology, unmet needs, or treatment outcomes in patients with HER2 + metastatic breast cancer and BM., Results: Key clinical trials of HER2-targeting treatments for HER2 + metastatic breast cancer had varying eligibility criteria relating to BM, with only two trials-HER2CLIMB and DEBBRAH-including patients with both active and stable BM. We also observed variance across assessed central nervous system (CNS)-focused endpoints (CNS objective response rate vs CNS progression-free survival vs time to CNS progression) and robustness of statistical analysis (prespecified vs exploratory)., Conclusions: There is an unmet need for standardization of clinical trial design for patients with HER2 + metastatic breast cancer and BM, to aid the interpretation of the global treatment landscape and ensure patients with all types of BM can access effective treatments., Competing Interests: Declaration of Competing Interest Volkmar Müller discloses speaker honoraria from Amgen, AstraZeneca, Daiichi Sankyo, Eisai, GSK, Pfizer, MSD, Medac, Novartis, Roche, Teva, Seagen, Onkowissen, high5 Oncology, Medscape, Gilead, Pierre Fabre; consultancy honoraria from Hexal, Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi Sankyo, Eisai, Lilly, Sanofi, Seagen, Gilead; institutional research support from Novartis, Roche, Seagen, Genentech; travel grants from Roche, Pfizer, Daiichi Sankyo, Gilead. Rupert Bartsch discloses advisory roles for AstraZeneca, Daiichi Sankyo, Eisai, Eli-Lilly, Gilead, Gruenenthal, MSD, Novartis, Pfizer, Pierre-Fabre, Puma, Roche, Seagen; lecture honoraria for Astra-Zeneca, Daiichi Sankyo, Eisai, Eli-Lilly, Gilead, Gruenenthal, MSD, Novartis, Pfizer, Pierre-Fabre, Roche, Seagen; research support for Daiichi Sankyo, MSD, Novartis, Roche. Nancy U. Lin discloses institutional research support for Genentech, Pfizer, Merck, Seagen, Zion Pharmaceuticals, Olema Pharmaceuticals, AstraZeneca; consulting honoraria for Puma, Seagen, Daichii-Sankyo, AstraZeneca, Denali Therapeutics, Prelude Therapeutics, Olema Pharmaceuticals, Aleta BioPharma, Affinia Therapeutics, Voyager Therapeutics, Janssen; stock and other ownership interests in Artera Inc. (<$50k and <5% as it relates to consulting activities; options are not currently valued or in-hand); royalties – up to date (book). Filippo Montemurro discloses consulting or advisory roles for AstraZeneca, Eli Lilly, Roche, Novartis, Seagen, Pfizer, MSD, Daiichi Sankyo and Pierre Fabre. Mark D. Pegram discloses consulting or advisory role for Roche/Genentech, AstraZeneca/Daiichi Sankyo, Seagen. Sara M. Tolaney discloses consulting or advisory roles for Novartis, Pfizer, Merck, Lilly, Nektar, NanoString Technologies, AstraZeneca, Puma Biotechnology, Genentech/Roche, Eisai, Sanofi, Bristol Myers Squibb, Seagen, Odonate Therapeutics, OncoPep, Kyowa Hakko Kirin, Samsung Bioepis, CytomX Therapeutics, Daiichi Sankyo, Athenex, Gilead, Mersana, Certara, Chugai Pharma, Ellipses Pharma, Infinity, 4D Pharma, OncoSec Medical Inc., BeyondSpring Pharmaceuticals, OncXerna, Zymeworks, Zentalis, Blueprint Medicines, Reveal Genomics, ARC Therapeutics; research funding for Genentech/Roche, Merck, Exelixis, Pfizer, Lilly, Novartis, Bristol Myers Squibb, Eisai, AstraZeneca, NanoString Technologies, Cyclacel, Nektar, Gilead, Odonate Therapeutics, Sanofi, Seagen., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Margetuximab Versus Trastuzumab in Patients With Previously Treated HER2-Positive Advanced Breast Cancer (SOPHIA): Final Overall Survival Results From a Randomized Phase 3 Trial.
- Author
-
Rugo HS, Im SA, Cardoso F, Cortes J, Curigliano G, Musolino A, Pegram MD, Bachelot T, Wright GS, Saura C, Escrivá-de-Romaní S, De Laurentiis M, Schwartz GN, Pluard TJ, Ricci F, Gwin WR 3rd, Levy C, Brown-Glaberman U, Ferrero JM, de Boer M, Kim SB, Petráková K, Yardley DA, Freedman O, Jakobsen EH, Gal-Yam EN, Yerushalmi R, Fasching PA, Kaufman PA, Ashley EJ, Perez-Olle R, Hong S, Rosales MK, and Gradishar WJ
- Subjects
- Humans, Female, Trastuzumab adverse effects, Receptor, ErbB-2, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms drug therapy, Breast Neoplasms genetics
- Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported. Final overall survival (OS) in SOPHIA (ClinicalTrials.gov identifier: NCT02492711), a study of margetuximab versus trastuzumab, both with chemotherapy, in patients with previously treated human epidermal growth factor receptor 2-positive advanced breast cancer, is reported with updated safety. Overall, 536 patients in the intention-to-treat population were randomly assigned to margetuximab (15 mg/kg intravenously once every 3 weeks; n = 266) plus chemotherapy or trastuzumab (6 mg/kg intravenously once every 3 weeks after a loading dose of 8 mg/kg; n = 270) plus chemotherapy. Primary end points were progression-free survival, previously reported, and OS. Final OS analysis was triggered by 385 prespecified events. The median OS was 21.6 months (95% CI, 18.89 to 25.07) with margetuximab versus 21.9 months (95% CI, 18.69 to 24.18) with trastuzumab (hazard ratio [HR], 0.95; 95% CI, 0.77 to 1.17; P = .620). Preplanned, exploratory analysis of CD16A genotyping suggested a possible improvement in OS for margetuximab in CD16A-158FF patients versus trastuzumab (median OS, 23.6 v 19.2 months; HR, 0.72; 95% CI, 0.52 to 1.00) and a possible improvement in OS for trastuzumab in CD16A-158VV patients versus margetuximab (median OS, 31.1 v 22.0 months; HR, 1.77; 95% CI, 1.01 to 3.12). Margetuximab safety was comparable with trastuzumab. Final overall OS analysis did not demonstrate margetuximab advantage over trastuzumab. Margetuximab studies in patients with human epidermal growth factor receptor 2-positive breast cancer with different CD16A allelic variants are warranted.
- Published
- 2023
- Full Text
- View/download PDF
15. Editorial: Metabolic Abnormalities and Breast Cancer: Challenges From Bench to Bedside.
- Author
-
Wang Z, Li P, Pegram MD, and Chen X
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
16. The Phase II MutHER Study of Neratinib Alone and in Combination with Fulvestrant in HER2-Mutated, Non-amplified Metastatic Breast Cancer.
- Author
-
Ma CX, Luo J, Freedman RA, Pluard TJ, Nangia JR, Lu J, Valdez-Albini F, Cobleigh M, Jones JM, Lin NU, Winer EP, Marcom PK, Thomas S, Anderson J, Haas B, Bucheit L, Bryce R, Lalani AS, Carey LA, Goetz MP, Gao F, Kimmick G, Pegram MD, Ellis MJ, and Bose R
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Female, Fulvestrant, Humans, Quinolines, Receptor, ErbB-2 genetics, Receptor, ErbB-2 therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Purpose: HER2 mutations (HER2mut) induce endocrine resistance in estrogen receptor-positive (ER+) breast cancer., Patients and Methods: In this single-arm multi-cohort phase II trial, we evaluated the efficacy of neratinib plus fulvestrant in patients with ER+/HER2mut, HER2 non-amplified metastatic breast cancer (MBC) in the fulvestrant-treated (n = 24) or fulvestrant-naïve cohort (n = 11). Patients with ER-negative (ER-)/HER2mut MBC received neratinib monotherapy in an exploratory ER- cohort (n = 5)., Results: The clinical benefit rate [CBR (95% confidence interval)] was 38% (18%-62%), 30% (7%-65%), and 25% (1%-81%) in the fulvestrant-treated, fulvestrant-naïve, and ER- cohorts, respectively. Adding trastuzumab at progression in 5 patients resulted in three partial responses and one stable disease ≥24 weeks. CBR appeared positively associated with lobular histology and negatively associated with HER2 L755 alterations. Acquired HER2mut were detected in 5 of 23 patients at progression., Conclusions: Neratinib and fulvestrant are active for ER+/HER2mut MBC. Our data support further evaluation of dual HER2 blockade for the treatment of HER2mut MBC., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. Role of Fcγ receptors in HER2-targeted breast cancer therapy.
- Author
-
Musolino A, Gradishar WJ, Rugo HS, Nordstrom JL, Rock EP, Arnaldez F, and Pegram MD
- Subjects
- Breast Neoplasms mortality, Female, Humans, Retrospective Studies, Survival Analysis, Adaptive Immunity immunology, Breast Neoplasms genetics, Immunity, Innate immunology, Receptor, ErbB-2 metabolism, Receptors, IgG metabolism
- Abstract
Several therapeutic monoclonal antibodies (mAbs), including those targeting epidermal growth factor receptor, human epidermal growth factor receptor 2 (HER2), and CD20, mediate fragment crystallizable gamma receptor (FcγR)-dependent activities as part of their mechanism of action. These activities include induction of antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), which are innate immune mechanisms of cancer cell elimination. FcγRs are distinguished by their affinity for the Fc fragment, cell distribution, and type of immune response they induce. Activating FcγRIIIa (CD16A) on natural killer cells plays a crucial role in mediating ADCC, and activating FcγRIIa (CD32A) and FcγRIIIa on macrophages are important for mediating ADCP. Polymorphisms in FcγRIIIa and FcγRIIa generate variants that bind to the Fc portion of antibodies with different affinities. This results in differential FcγR-mediated activities associated with differential therapeutic outcomes across multiple clinical settings, from early stage to metastatic disease, in patients with HER2+ breast cancer treated with the anti-HER2 mAb trastuzumab. Trastuzumab has, nonetheless, revolutionized HER2+ breast cancer treatment, and several HER2-directed mAbs have been developed using Fc glyco-engineering or Fc protein-engineering to enhance FcγR-mediated functions. An example of an approved anti-HER2 Fc-engineered chimeric mAb is margetuximab, which targets the same epitope as trastuzumab, but features five amino acid substitutions in the IgG 1 Fc domain that were deliberately introduced to increase binding to activating FcγRIIIa and decrease binding to inhibitory FcγRIIb (CD32B). Margetuximab enhances Fc-dependent ADCC in vitro more potently than the combination of pertuzumab (another approved mAb directed against an alternate HER2 epitope) and trastuzumab. Margetuximab administration also enhances HER2-specific B cell and T cell-mediated responses ex vivo in samples from patients treated with prior lines of HER2 antibody-based therapies. Stemming from these observations, a worthwhile future goal in the treatment of HER2+ breast cancer is to promote combinatorial approaches that better eradicate HER2+ cancer cells via enhanced immunological mechanisms., Competing Interests: Competing interests: AM reports grants from Roche and Eisai; personal fees from MacroGenics, Roche, Eisai, Novartis, and Lilly; participation in advisory boards from MacroGenics, Roche, Eisai, Novartis, Lilly. WJG has nothing to disclose. HSR reports personal fees for short-term consulting from Puma and Samsung; institutional grants for clinical research study activities from MacroGenics, Roche, Pfizer, Novartis, Lilly, Merck, Seattle Genetics, Odonate Therapeutics, Eisai, Sermonix, and Immunomedics, Daiichi Sankyo. MDP reports personal consulting fees from MacroGenics, AstraZeneca/Daiichi Sankyo, Pfizer, and Roche/Genentech, and grant support on this topic from the Parker Institute for Cancer Immunotherapy and the Mary Kay Foundation. JLN is an employee of MacroGenics. EPR was an employee of MacroGenics and is now an employee of Partner Therapeutics. FA was an employee of MacroGenics and is now an employee of AstraZeneca., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
18. Reply to J. Wei et al.
- Author
-
Lin NU, Pegram M, Sahebjam S, Ibrahim N, Fung A, Cheng A, Nicholas A, Kirschbrown W, and Kumthekar P
- Abstract
Competing Interests: Nancy U. LinConsulting or Advisory Role: Seattle Genetics, Puma Biotechnology, Daiichi Sankyo, California Institute for Regenerative Medicine (CIRM), Denali Therapeutics, AstraZeneca, Prelude TherapeuticsResearch Funding: Genentech, Pfizer, Seattle Genetics, MerckPatents, Royalties, Other Intellectual Property: Royalties for chapter in Up-to-Date regarding management of breast cancer brain metastases, Royalties, Jones & Bartlett, Mark PegramHonoraria: Genentech/Roche, Pfizer¸ Seattle GeneticsConsulting or Advisory Role: Genentech/Roche, Pfizer, Seattle Genetics Solmaz SahebjamStock and Other Ownership Interests: Allergan, AbbVieConsulting or Advisory Role: Merck, Boehringer IngelheimResearch Funding: Bristol Myers Squibb, Brooklyn ImmunoTherapeutics, MerckTravel, Accommodations, Expenses: Lilly Nuhad IbrahimHonoraria: Roche, Novartis, Athenex¸ ImmunomedicsConsulting or Advisory Role: ImmunomedicsSpeakers' Bureau: Roche, NovartisTravel, Accommodations, Expenses: Roche, Novartis Anita FungEmployment: Genentech/RocheStock and Other Ownership Interests: Genentech/Roche Anna ChengEmployment: GenentechStock and Other Ownership Interests: Roche, Calithera Biosciences Alan NicholasEmployment: GenentechStock and Other Ownership Interests: Genentech Whitney KirschbrownEmployment: Genentech/RocheStock and Other Ownership Interests: Genentech/RochePatents, Royalties, Other Intellectual Property: Inventor on a pertuzumab-related patent filing Priya KumthekarStock and Other Ownership Interests: Vivacitas OncologyConsulting or Advisory Role: Orbus Therapeutics, Janssen, Novocure¸ ElevateBio, BioceptResearch Funding: Novocure, Orbus Therapeutics, DNAtrix, Genentech, AngiochemPatents, Royalties, Other Intellectual Property: Utility patent for ANG 1005. No financial gain, no royaltiesNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
19. Real-world Evidence of Diagnostic Testing and Treatment Patterns in US Patients With Breast Cancer With Implications for Treatment Biomarkers From RNA Sequencing Data.
- Author
-
Fernandes LE, Epstein CG, Bobe AM, Bell JSK, Stumpe MC, Salazar ME, Salahudeen AA, Pe Benito RA, McCarter C, Leibowitz BD, Kase M, Igartua C, Huether R, Hafez A, Beaubier N, Axelson MD, Pegram MD, Sammons SL, O'Shaughnessy JA, and Palmer GA
- Subjects
- Aged, Breast Neoplasms therapy, Databases, Factual, Feasibility Studies, Female, Gene Expression Profiling, Humans, Longitudinal Studies, Middle Aged, Receptor, ErbB-2 genetics, Receptors, Estrogen genetics, Retrospective Studies, Sensitivity and Specificity, United States, Breast Neoplasms diagnosis, Breast Neoplasms genetics, Sequence Analysis, RNA
- Abstract
Objective/background: We performed a retrospective analysis of longitudinal real-world data (RWD) from patients with breast cancer to replicate results from clinical studies and demonstrate the feasibility of generating real-world evidence. We also assessed the value of transcriptome profiling as a complementary tool for determining molecular subtypes., Methods: De-identified, longitudinal data were analyzed after abstraction from records of patients with breast cancer in the United States (US) structured and stored in the Tempus database. Demographics, clinical characteristics, molecular subtype, treatment history, and survival outcomes were assessed according to strict qualitative criteria. RNA sequencing and clinical data were used to predict molecular subtypes and signaling pathway enrichment., Results: The clinical abstraction cohort (n = 4000) mirrored the demographics and clinical characteristics of patients with breast cancer in the US, indicating feasibility for RWE generation. Among patients who were human epidermal growth factor receptor 2-positive (HER2
+ ), 74.2% received anti-HER2 therapy, with ∼70% starting within 3 months of a positive test result. Most non-treated patients were early stage. In this RWD set, 31.7% of patients with HER2+ immunohistochemistry (IHC) had discordant fluorescence in situ hybridization results recorded. Among patients with multiple HER2 IHC results at diagnosis, 18.6% exhibited intra-test discordance. Through development of a whole-transcriptome model to predict IHC receptor status in the molecular sequenced cohort (n = 400), molecular subtypes were resolved for all patients (n = 36) with equivocal HER2 statuses from abstracted test results. Receptor-related signaling pathways were differentially enriched between clinical molecular subtypes., Conclusions: RWD in the Tempus database mirrors the overall population of patients with breast cancer in the US. These results suggest that real-time, RWD analyses are feasible in a large, highly heterogeneous database. Furthermore, molecular data may aid deficiencies and discrepancies observed from breast cancer RWD., (Copyright © 2020. Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
20. First-in-Human, Phase 1 Dose-Escalation Study of Biparatopic Anti-HER2 Antibody-Drug Conjugate MEDI4276 in Patients with HER2-positive Advanced Breast or Gastric Cancer.
- Author
-
Pegram MD, Hamilton EP, Tan AR, Storniolo AM, Balic K, Rosenbaum AI, Liang M, He P, Marshall S, Scheuber A, Das M, and Patel MR
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Follow-Up Studies, Maximum Tolerated Dose, Prognosis, Survival Rate, Tissue Distribution, Antibodies, Monoclonal chemistry, Antineoplastic Agents, Immunological chemistry, Antineoplastic Agents, Immunological pharmacokinetics, Antineoplastic Agents, Immunological pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms immunology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Immunoconjugates chemistry, Immunoconjugates pharmacokinetics, Immunoconjugates pharmacology, Receptor, ErbB-2 immunology, Stomach Neoplasms drug therapy, Stomach Neoplasms immunology, Stomach Neoplasms metabolism, Stomach Neoplasms pathology
- Abstract
MEDI4276 is a biparatopic tetravalent antibody targeting two nonoverlapping epitopes in subdomains 2 and 4 of the HER2 ecto-domain, with site-specific conjugation to a tubulysin-based microtubule inhibitor payload. MEDI4276 demonstrates enhanced cellular internalization and cytolysis of HER2-positive tumor cells in vitro This was a first-in-human, dose-escalation clinical trial in patients with HER2-positive advanced or metastatic breast cancer or gastric cancer. MEDI4276 doses escalated from 0.05 to 0.9 mg/kg (60- to 90-minute intravenous infusion every 3 weeks). Primary endpoints were safety and tolerability; secondary endpoints included antitumor activity (objective response, progression-free survival, and overall survival), pharmacokinetics, and immunogenicity. Forty-seven patients (median age 59 years; median of seven prior treatment regimens) were treated. The maximum tolerated dose was exceeded at 0.9 mg/kg with two patients experiencing dose-limiting toxicities (DLTs) of grade 3 liver function test (LFT) increases, one of whom also had grade 3 diarrhea, which resolved. Two additional patients reported DLTs of grade 3 LFT increases at lower doses (0.4 and 0.6 mg/kg). The most common (all grade) drug-related adverse events (AEs) were nausea (59.6%), fatigue (44.7%), aspartate aminotransferase (AST) increased (42.6%), and vomiting (38.3%). The most common grade 3/4 drug-related AE was AST increased (21.3%). Five patients had drug-related AEs leading to treatment discontinuation. In the as-treated population, there was one complete response (0.5 mg/kg; breast cancer), and two partial responses (0.6 and 0.75 mg/kg; breast cancer)-all had prior trastuzumab, pertuzumab, and ado-trastuzumab emtansine (T-DM1). MEDI4276 has demonstrable clinical activity but displays intolerable toxicity at doses >0.3 mg/kg., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
21. Single-cell immunoblotting resolves estrogen receptor-α isoforms in breast cancer.
- Author
-
Kim JJ, Liang W, Kang CC, Pegram MD, and Herr AE
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Estrogen Receptor alpha genetics, Female, Humans, Immunoblotting, Phosphorylation drug effects, Principal Component Analysis, Protein Isoforms genetics, Protein Isoforms metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Signal Transduction genetics, Single-Cell Analysis instrumentation, Tamoxifen pharmacology, Estrogen Receptor alpha metabolism, Single-Cell Analysis methods
- Abstract
An array of isoforms of the nuclear estrogen receptor alpha (ER-α) protein contribute to heterogeneous response in breast cancer (BCa); yet, a single-cell analysis tool that distinguishes the full-length ER-α66 protein from the activation function-1 deficient ER-α46 isoform has not been reported. Specific detection of protein isoforms is a gap in single-cell analysis tools, as the de facto standard immunoassay requires isoform-specific antibody probes. Consequently, to scrutinize hormone response heterogeneity among BCa tumor cells, we develop a precision tool to specifically measure ER-α66, ER- α46, and eight ER-signaling proteins with single-cell resolution in the highly hetero-clonal MCF-7 BCa cell line. With a literature-validated pan-ER immunoprobe, we distinguish ER-α66 from ER-α46 in each individual cell. We identify ER-α46 in 5.5% of hormone-sensitive (MCF-7) and 4.2% of hormone-insensitive (MDA-MB-231) BCa cell lines. To examine whether the single-cell immunoblotting can capture cellular responses to hormones, we treat cells with tamoxifen and identify different sub-populations of ER-α46: (i) ER-α46 induces phospho-AKT at Ser473, (ii) S6-ribosomal protein, an upstream ER target, activates both ER-α66 and ER-α46 in MCF-7 cells, and (iii) ER-α46 partitions MDA-MB-231 subpopulations, which are responsive to tamoxifen. Unlike other single-cell immunoassays, multiplexed single-cell immunoblotting reports-in the same cell-tamoxifen effects on ER signaling proteins and on distinct isoforms of the ER-α protein., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: C.-C.K., and A.E.H. are co-inventors on intellectual property related to the single-cell immunoblot described here and may benefit from royalties from licensing. A.E.H. has financial interest in commercialization efforts. The competing interests do not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
22. Combining CD47 blockade with trastuzumab eliminates HER2-positive breast cancer cells and overcomes trastuzumab tolerance.
- Author
-
Upton R, Banuelos A, Feng D, Biswas T, Kao K, McKenna K, Willingham S, Ho PY, Rosental B, Tal MC, Raveh T, Volkmer JP, Pegram MD, and Weissman IL
- Subjects
- Animals, Antibody-Dependent Cell Cytotoxicity drug effects, Breast Neoplasms genetics, Breast Neoplasms immunology, CD47 Antigen antagonists & inhibitors, CD47 Antigen genetics, Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Therapy, Combination, Female, Humans, Immunotherapy, Macrophages drug effects, Macrophages immunology, Mice, Mice, Inbred C57BL, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents, Immunological administration & dosage, Breast Neoplasms drug therapy, CD47 Antigen immunology, Trastuzumab administration & dosage
- Abstract
Trastuzumab, a targeted anti-human epidermal-growth-factor receptor-2 (HER2) monoclonal antibody, represents a mainstay in the treatment of HER2-positive (HER2
+ ) breast cancer. Although trastuzumab treatment is highly efficacious for early-stage HER2+ breast cancer, the majority of advanced-stage HER2+ breast cancer patients who initially respond to trastuzumab acquire resistance to treatment and relapse, despite persistence of HER2 gene amplification/overexpression. Here, we sought to leverage HER2 overexpression to engage antibody-dependent cellular phagocytosis (ADCP) through a combination of trastuzumab and anti-CD47 macrophage checkpoint immunotherapy. We have previously shown that blockade of CD47, a surface protein expressed by many malignancies (including HER2+ breast cancer), is an effective anticancer therapy. CD47 functions as a "don't eat me" signal through its interaction with signal regulatory protein-α (SIRPα) on macrophages to inhibit phagocytosis. Hu5F9-G4 (magrolimab), a humanized monoclonal antibody against CD47, blocks CD47's "don't eat me" signal, thereby facilitating macrophage-mediated phagocytosis. Preclinical studies have shown that combining Hu5F9-G4 with tumor-targeting antibodies, such as rituximab, further enhances Hu5F9-G4's anticancer effects via ADCP. Clinical trials have additionally demonstrated that Hu5F9-G4, in combination with rituximab, produced objective responses in patients whose diffuse large B cell lymphomas had developed resistance to rituximab and chemotherapy. These studies led us to hypothesize that combining Hu5F9-G4 with trastuzumab would produce an anticancer effect in antibody-dependent cellular cytotoxicity (ADCC)-tolerant HER2+ breast cancer. This combination significantly suppressed the growth of ADCC-tolerant HER2+ breast cancers via Fc-dependent ADCP. Our study demonstrates that combining trastuzumab and Hu5F9-G4 represents a potential new treatment option for HER2+ breast cancer patients, even for patients whose tumors have progressed after trastuzumab., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
23. Efficacy of Margetuximab vs Trastuzumab in Patients With Pretreated ERBB2-Positive Advanced Breast Cancer: A Phase 3 Randomized Clinical Trial.
- Author
-
Rugo HS, Im SA, Cardoso F, Cortés J, Curigliano G, Musolino A, Pegram MD, Wright GS, Saura C, Escrivá-de-Romaní S, De Laurentiis M, Levy C, Brown-Glaberman U, Ferrero JM, de Boer M, Kim SB, Petráková K, Yardley DA, Freedman O, Jakobsen EH, Kaufman B, Yerushalmi R, Fasching PA, Nordstrom JL, Bonvini E, Koenig S, Edlich S, Hong S, Rock EP, and Gradishar WJ
- Subjects
- Ado-Trastuzumab Emtansine, Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols, Female, Humans, Middle Aged, Receptor, ErbB-2 analysis, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal therapeutic use, Breast Neoplasms drug therapy, Trastuzumab adverse effects, Trastuzumab therapeutic use
- Abstract
Importance: ERRB2 (formerly HER2)-positive advanced breast cancer (ABC) remains typically incurable with optimal treatment undefined in later lines of therapy. The chimeric antibody margetuximab shares ERBB2 specificity with trastuzumab but incorporates an engineered Fc region to increase immune activation., Objective: To compare the clinical efficacy of margetuximab vs trastuzumab, each with chemotherapy, in patients with pretreated ERBB2-positive ABC., Design, Setting, and Participants: The SOPHIA phase 3 randomized open-label trial of margetuximab plus chemotherapy vs trastuzumab plus chemotherapy enrolled 536 patients from August 26, 2015, to October 10, 2018, at 166 sites in 17 countries. Eligible patients had disease progression on 2 or more prior anti-ERBB2 therapies and 1 to 3 lines of therapy for metastatic disease. Data were analyzed from February 2019 to October 2019., Interventions: Investigators selected chemotherapy before 1:1 randomization to margetuximab, 15 mg/kg, or trastuzumab, 6 mg/kg (loading dose, 8 mg/kg), each in 3-week cycles. Stratification factors were metastatic sites (≤2, >2), lines of therapy (≤2, >2), and chemotherapy choice., Main Outcomes and Measures: Sequential primary end points were progression-free survival (PFS) by central blinded analysis and overall survival (OS). All α was allocated to PFS, followed by OS. Secondary end points were investigator-assessed PFS and objective response rate by central blinded analysis., Results: A total of 536 patients were randomized to receive margetuximab (n = 266) or trastuzumab (n = 270). The median age was 56 (27-86) years; 266 (100%) women were in the margetuximab group, while 267 (98.9%) women were in the trastuzumab group. Groups were balanced. All but 1 patient had received prior pertuzumab, and 489 (91.2%) had received prior ado-trastuzumab emtansine. Margetuximab improved primary PFS over trastuzumab with 24% relative risk reduction (hazard ratio [HR], 0.76; 95% CI, 0.59-0.98; P = .03; median, 5.8 [95% CI, 5.5-7.0] months vs 4.9 [95% CI, 4.2-5.6] months; October 10, 2018). After the second planned interim analysis of 270 deaths, median OS was 21.6 months with margetuximab vs 19.8 months with trastuzumab (HR, 0.89; 95% CI, 0.69-1.13; P = .33; September 10, 2019), and investigator-assessed PFS showed 29% relative risk reduction favoring margetuximab (HR, 0.71; 95% CI, 0.58-0.86; P < .001; median, 5.7 vs 4.4 months; September 10, 2019). Margetuximab improved objective response rate over trastuzumab: 22% vs 16% (P = .06; October 10, 2018), and 25% vs 14% (P < .001; September 10, 2019). Incidence of infusion-related reactions, mostly in cycle 1, was higher with margetuximab (35 [13.3%] vs 9 [3.4%]); otherwise, safety was comparable., Conclusions and Relevance: In this phase 3 randomized clinical trial, margetuximab plus chemotherapy had acceptable safety and a statistically significant improvement in PFS compared with trastuzumab plus chemotherapy in ERBB2-positive ABC after progression on 2 or more prior anti-ERBB2 therapies. Final OS analysis is expected in 2021., Trial Registration: ClinicalTrials.gov Identifier: NCT02492711.
- Published
- 2021
- Full Text
- View/download PDF
24. Mitochondrial copper depletion suppresses triple-negative breast cancer in mice.
- Author
-
Cui L, Gouw AM, LaGory EL, Guo S, Attarwala N, Tang Y, Qi J, Chen YS, Gao Z, Casey KM, Bazhin AA, Chen M, Hu L, Xie J, Fang M, Zhang C, Zhu Q, Wang Z, Giaccia AJ, Gambhir SS, Zhu W, Felsher DW, Pegram MD, Goun EA, Le A, and Rao J
- Subjects
- Animals, Cell Death, Cell Line, Tumor, Chelating Agents metabolism, Disease Models, Animal, Female, Humans, Mice, Oxidative Phosphorylation, Triple Negative Breast Neoplasms metabolism, Copper metabolism, Mitochondria metabolism, Triple Negative Breast Neoplasms pathology
- Abstract
Depletion of mitochondrial copper, which shifts metabolism from respiration to glycolysis and reduces energy production, is known to be effective against cancer types that depend on oxidative phosphorylation. However, existing copper chelators are too toxic or ineffective for cancer treatment. Here we develop a safe, mitochondria-targeted, copper-depleting nanoparticle (CDN) and test it against triple-negative breast cancer (TNBC). We show that CDNs decrease oxygen consumption and oxidative phosphorylation, cause a metabolic switch to glycolysis and reduce ATP production in TNBC cells. This energy deficiency, together with compromised mitochondrial membrane potential and elevated oxidative stress, results in apoptosis. CDNs should be less toxic than existing copper chelators because they favorably deprive copper in the mitochondria in cancer cells instead of systemic depletion. Indeed, we demonstrate low toxicity of CDNs in healthy mice. In three mouse models of TNBC, CDN administration inhibits tumor growth and substantially improves survival. The efficacy and safety of CDNs suggest the potential clinical relevance of this approach.
- Published
- 2021
- Full Text
- View/download PDF
25. A Novel HER2-targeted Antibody-drug Conjugate Offers the Possibility of Clinical Dosing at Trastuzumab-equivalent Exposure Levels.
- Author
-
Barfield RM, Kim YC, Chuprakov S, Zhang F, Bauzon M, Ogunkoya AO, Yeo D, Hickle C, Pegram MD, Rabuka D, and Drake PM
- Subjects
- Ado-Trastuzumab Emtansine adverse effects, Ado-Trastuzumab Emtansine pharmacokinetics, Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Female, Humans, Immunoconjugates adverse effects, Immunoconjugates chemistry, Immunoconjugates pharmacokinetics, Macaca fascicularis, Maximum Tolerated Dose, Rats, Rats, Sprague-Dawley, Receptor, ErbB-2 metabolism, Trastuzumab pharmacology, Xenograft Model Antitumor Assays, Ado-Trastuzumab Emtansine administration & dosage, Breast Neoplasms drug therapy, Immunoconjugates administration & dosage, Maytansine chemistry, Trastuzumab chemistry
- Abstract
Trastuzumab and the related ADC, ado-trastuzumab emtansine (T-DM1), both target HER2-overexpressing cells. Together, these drugs have treatment indications in both early-stage and metastatic settings for HER2
+ breast cancer. T-DM1 retains the antibody functionalities of trastuzumab and adds the potency of a cytotoxic maytansine payload. Interestingly, in the clinic, T-DM1 cannot always replace the use of trastuzumab plus chemotherapy administered together as single agents. We hypothesize that this failure may be due, in part, to the limited systemic exposure achieved by T-DM1 relative to trastuzumab because of toxicity-related dosing constraints on the ADC. We have developed a trastuzumab-based ADC site specifically conjugated to maytansine through a noncleavable linker. This construct, termed CAT-01-106, has a drug-to-antibody ratio (DAR) of 1.8, approximately half the average DAR of T-DM1, which comprises a mixture of antibodies variously conjugated with DARs ranging from 0 to 8. The high DAR species present in T-DM1 contribute to its toxicity and limit its clinical dose. CAT-01-106 showed superior in vivo efficacy compared with T-DM1 at equal payload dosing and was equally or better tolerated compared with T-DM1 at equal payload dosing up to 120 mg/kg in Sprague-Dawley rats and 60 mg/kg in cynomolgus monkeys. CAT-01-106 also showed improved pharmacokinetics in rats relative to T-DM1, with 40% higher ADC exposure levels. Together, the data suggest that CAT-01-106 may be sufficiently tolerable to enable clinical dosing at trastuzumab-equivalent exposure levels, combining the functions of both the antibody and the payload in one drug and potentially improving patient outcomes., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
26. Extracellular Vesicle-Mediated In Vitro Transcribed mRNA Delivery for Treatment of HER2 + Breast Cancer Xenografts in Mice by Prodrug CB1954 without General Toxicity.
- Author
-
Forterre AV, Wang JH, Delcayre A, Kim K, Green C, Pegram MD, Jeffrey SS, and Matin AC
- Subjects
- Animals, Apoptosis, Bacterial Proteins metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Proliferation, Extracellular Vesicles genetics, Female, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, ErbB-2 metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Bacterial Proteins genetics, Breast Neoplasms therapy, Extracellular Vesicles metabolism, Gene Transfer Techniques, Genetic Therapy, Prodrugs pharmacology, RNA, Messenger administration & dosage
- Abstract
Prodrugs are harmless until activated by a bacterial or viral gene product; they constitute the basis of gene-delivered prodrug therapies called GDEPT, which can kill tumors without major side effects. Previously, we utilized the prodrug CNOB (C
16 H7 CIN2 O4 ; not clinically tested) and enzyme HChrR6 in GDEPT to generate the drug MCHB (C16 H9 CIN2 O2 ) in tumors. Extracellular vesicles (EVs) were used for directed gene delivery and HChrR6 mRNA as gene. Here, the clinical transfer of this approach is enhanced by: (i) use of CB1954 (tretazicar) for which safe human dose is established; HChrR6 can activate this prodrug. (ii) EVs delivered in vitro transcribed (IVT) HChrR6 mRNA, eliminating the potentially harmful plasmid transfection of EV producer cells we utilized previously; this has not been done before. IVT mRNA loading of EVs required several steps. Naked mRNA being unstable, we ensured its prodrug activating functionality at each step. This was not possible using tretazicar itself; we relied instead on HChrR6's ability to convert CNOB into MCHB, whose fluorescence is easily visualizable. HChrR6 mRNA-translated product's ability to generate fluorescence from CNOB vicariously indicated its competence for tretazicar activation. (iii) Systemic IVT mRNA-loaded EVs displaying an anti-HER2 single-chain variable fragment ("IVT EXO-DEPTs") and tretazicar caused growth arrest of human HER2+ breast cancer xenografts in athymic mice. As this occurred without injury to other tissues, absence of off-target mRNA delivery is strongly indicated. Many cancer sites are not amenable for direct gene injection, but current GDEPTs require this. In circumventing this need, a major advance in GDEPT applicability has been accomplished., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
27. HER2-Overexpressing/Amplified Breast Cancer as a Testing Ground for Antibody-Drug Conjugate Drug Development in Solid Tumors.
- Author
-
Pegram MD, Miles D, Tsui CK, and Zong Y
- Subjects
- Antineoplastic Agents, Immunological therapeutic use, Breast Neoplasms enzymology, Breast Neoplasms immunology, Breast Neoplasms pathology, Clinical Trials as Topic, Drug Development, Drug Resistance, Neoplasm, Female, Gene Amplification, Humans, Molecular Targeted Therapy methods, Randomized Controlled Trials as Topic, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology, Tissue Distribution, Ado-Trastuzumab Emtansine therapeutic use, Breast Neoplasms drug therapy, Immunoconjugates pharmacokinetics, Immunoconjugates therapeutic use, Receptor, ErbB-2 biosynthesis
- Abstract
Efficacy data from the KATHERINE clinical trial, comparing the HER2-directed antibody-drug conjugate (ADC) ado-trastuzumab emtansine (T-DM1) to trastuzumab in patients with early-stage HER2-amplified/overexpressing breast cancer with residual disease after neoadjuvant therapy, demonstrates superiority of T-DM1 (HR for invasive disease or death, 0.50; P < 0.001). This establishes foundational precedent for ADCs as effective therapy for treatment of subclinical micrometastasis in an adjuvant (or post-neoadjuvant) early-stage solid tumor setting. Despite this achievement, general principles from proposed systems pharmacokinetic modeling for intracellular processing of ADCs indicate potential shortcomings of T-DM1: (i) C
max limited by toxicities; (ii) slow internalization rate; (iii) resistance mechanisms due to defects in intracellular trafficking [loss of lysosomal transporter solute carrier family 46 member 3, (SLC46A3)], and increased expression of drug transporters MDR1 and MRP1; and (iv) lack of payload bystander effects limiting utility in tumors with heterogeneous HER2 expression. These handicaps may explain the inferiority of T-DM1-based therapy in the neoadjuvant and first-line metastatic HER2+ breast cancer settings, and lack of superiority to chemotherapy in HER2+ advanced gastric cancer. In this review, we discuss how each of these limitations is being addressed by manipulating internalization and trafficking using HER2:HER2 bispecific or biparatopic antibody backbones, using site-specific, fixed DAR conjugation chemistry, and payload swapping to exploit alternative intracellular targets and to promote bystander effects. Newer HER2-directed ADCs have impressive clinical activity even against tumors with lower levels of HER2 receptor expression. Finally, we highlight ongoing clinical efforts to combine HER2 ADCs with other treatment modalities, including chemotherapy, molecularly targeted therapies, and immunotherapy., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
28. Induced pluripotent stem cells as a novel cancer vaccine.
- Author
-
Wang L, Pegram MD, and Wu JC
- Subjects
- Animals, Antigens, Neoplasm immunology, Cancer Vaccines classification, Humans, Induced Pluripotent Stem Cells cytology, Mice, Neoplasms immunology, Quality of Life, Cancer Vaccines therapeutic use, Immunotherapy methods, Induced Pluripotent Stem Cells transplantation, Neoplasms therapy
- Abstract
Introduction : Although many current cancer therapies are effective, the mortality rate globally is unacceptably high. Cancer remains the second leading cause of death worldwide after heart disease and has caused nearly 10 million deaths in 2018. Additionally, current preventive therapies for cancer are underdeveloped, undermining the quality of life of high-risk individuals. Therefore, new treatment options for targeting cancer are urgently needed. In a recent study, researchers adopted an autologous iPSC-based vaccine to present a broad spectrum of tumor antigens to the immune system and succeeded in orchestrating a strong prophylactic immunity towards multiple types of cancer in mice. Areas covered : In this review, we provide an overview of how cancer develops, the role of immune surveillance in cancer progression, the current status and challenges of cancer immunotherapy as well as the genetic overlap between pluripotent stem cells and cancer cells. Finally, we discuss the rationale for an autologous iPSC-based vaccine and its applications in murine cancer models. Expert opinion : The autologous iPSC-based vaccine is a promising preventive and therapeutic strategy for fighting various types of cancers. Continuing efforts and clinical/translational follow-up studies may bring an autologous iPSC-based cancer vaccination approach from bench to bedside.
- Published
- 2019
- Full Text
- View/download PDF
29. First-in-Human, First-in-Class Phase I Trial of the Anti-CD47 Antibody Hu5F9-G4 in Patients With Advanced Cancers.
- Author
-
Sikic BI, Lakhani N, Patnaik A, Shah SA, Chandana SR, Rasco D, Colevas AD, O'Rourke T, Narayanan S, Papadopoulos K, Fisher GA, Villalobos V, Prohaska SS, Howard M, Beeram M, Chao MP, Agoram B, Chen JY, Huang J, Axt M, Liu J, Volkmer JP, Majeti R, Weissman IL, Takimoto CH, Supan D, Wakelee HA, Aoki R, Pegram MD, and Padda SK
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized immunology, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological immunology, Antineoplastic Agents, Immunological pharmacokinetics, Biopsy, CD47 Antigen immunology, Cohort Studies, Female, Humans, Lymphoma immunology, Lymphoma metabolism, Lymphoma pathology, Male, Middle Aged, Neoplasms immunology, Neoplasms metabolism, Neoplasms pathology, Antibodies, Monoclonal, Humanized administration & dosage, Lymphoma drug therapy, Neoplasms drug therapy
- Abstract
Purpose: To evaluate the safety, pharmacokinetics, and pharmacodynamics of Hu5F9-G4 (5F9), a humanized IgG4 antibody that targets CD47 to enable phagocytosis., Patients and Methods: Adult patients with solid tumors were treated in four cohorts: part A, to determine a priming dose; part B, to determine a weekly maintenance dose; part C, to study a loading dose in week 2; and a tumor biopsy cohort., Results: Sixty-two patients were treated: 11 in part A, 14 in B, 22 in C, and 15 in the biopsy cohort. Part A used doses that ranged from 0.1 to 3 mg/kg. On the basis of tolerability and receptor occupancy studies that showed 100% CD47 saturation on RBCs, 1 mg/kg was selected as the priming dose. In subsequent groups, patients were treated with maintenance doses that ranged from 3 to 45 mg/kg, and most toxicities were mild to moderate. These included transient anemia (57% of patients), hemagglutination on peripheral blood smear (36%), fatigue (64%), headaches (50%), fever (45%), chills (45%), hyperbilirubinemia (34%), lymphopenia (34%), infusion-related reactions (34%), and arthralgias (18%). No maximum tolerated dose was reached with maintenance doses up to 45 mg/kg. At doses of 10 mg/kg or more, the CD47 antigen sink was saturated by 5F9, and a 5F9 half-life of approximately 13 days was observed. Strong antibody staining of tumor tissue was observed in a patient at 30 mg/kg. Two patients with ovarian/fallopian tube cancers had partial remissions for 5.2 and 9.2 months., Conclusion: 5F9 is well tolerated using a priming dose at 1 mg/kg on day 1 followed by maintenance doses of up to 45 mg/kg weekly.
- Published
- 2019
- Full Text
- View/download PDF
30. Clinical validation of an immunohistochemistry-based CanAssist-Breast test for distant recurrence prediction in hormone receptor-positive breast cancer patients.
- Author
-
Bakre MM, Ramkumar C, Attuluri AK, Basavaraj C, Prakash C, Buturovic L, Madhav L, Naidu N, R P, Somashekhar SP, Gupta S, Doval DC, and Pegram MD
- Subjects
- Adult, Aged, Algorithms, Breast Neoplasms pathology, Breast Neoplasms therapy, Female, Humans, Kaplan-Meier Estimate, Lymphatic Metastasis, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Neoplasm Staging, Prognosis, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Retrospective Studies, Risk Assessment methods, Support Vector Machine, Biomarkers, Tumor metabolism, Breast Neoplasms diagnosis
- Abstract
CanAssist-Breast (CAB) is an immunohistochemistry (IHC)-based prognostic test for early-stage Hormone Receptor (HR+)-positive breast cancer patients. CAB uses a Support Vector Machine (SVM) trained algorithm which utilizes expression levels of five biomarkers (CD44, ABCC4, ABCC11, N-Cadherin, and Pan-Cadherin) and three clinical parameters such as tumor size, grade, and node status as inputs to generate a risk score and categorizes patients as low- or high-risk for distant recurrence within 5 years of diagnosis. In this study, we present clinical validation of CAB. CAB was validated using a retrospective cohort of 857 patients. All patients were treated either with endocrine therapy or chemoendocrine therapy. Risk categorization by CAB was analyzed by calculating Distant Metastasis-Free Survival (DMFS) and recurrence rates using Kaplan-Meier survival curves. Multivariate analysis was performed to calculate Hazard ratios (HR) for CAB high-risk vs low-risk patients. The results showed that Distant Metastasis-Free Survival (DMFS) was significantly different (P-0.002) between low- (DMFS: 95%) and high-risk (DMFS: 80%) categories in the endocrine therapy treated alone subgroup (n = 195) as well as in the total cohort (n = 857, low-risk DMFS: 95%, high-risk DMFS: 84%, P < 0.0001). In addition, the segregation of the risk categories was significant (P = 0.0005) in node-positive patients, with a difference in DMFS of 12%. In multivariate analysis, CAB risk score was the most significant predictor of distant recurrence with hazard ratio of 3.2048 (P < 0.0001). CAB stratified patients into discrete risk categories with high statistical significance compared to Ki-67 and IHC4 score-based stratification. CAB stratified a higher percentage of the cohort (82%) as low-risk than IHC4 score (41.6%) and could re-stratify >74% of high Ki-67 and IHC4 score intermediate-risk zone patients into low-risk category. Overall the data suggest that CAB can effectively predict risk of distant recurrence with clear dichotomous high- or low-risk categorization., (© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
31. PF-05280014 (a trastuzumab biosimilar) plus paclitaxel compared with reference trastuzumab plus paclitaxel for HER2-positive metastatic breast cancer: a randomised, double-blind study.
- Author
-
Pegram MD, Bondarenko I, Zorzetto MMC, Hingmire S, Iwase H, Krivorotko PV, Lee KS, Li RK, Pikiel J, Aggarwal R, Ewesuedo R, Freyman A, Li R, Vana A, Yin D, Zacharchuk C, and Tan-Chiu E
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biosimilar Pharmaceuticals, Breast Neoplasms genetics, Breast Neoplasms pathology, Disease-Free Survival, Double-Blind Method, Female, Humans, Kaplan-Meier Estimate, Middle Aged, Neoplasm Metastasis, Paclitaxel administration & dosage, Paclitaxel adverse effects, Trastuzumab adverse effects, Trastuzumab chemistry, Antibodies, Monoclonal, Humanized administration & dosage, Breast Neoplasms drug therapy, Receptor, ErbB-2 genetics, Trastuzumab administration & dosage
- Abstract
Background: This randomised, double-blind study compared PF-05280014 (a trastuzumab biosimilar) with reference trastuzumab (Herceptin®) sourced from the European Union (trastuzumab-EU), when each was given with paclitaxel as first-line treatment for HER2-positive metastatic breast cancer., Methods: Between 4 April 2014 and 22 January 2016, 707 participants were randomised 1:1 to receive intravenous PF-05280014 plus paclitaxel (PF-05280014 group; n = 352) or trastuzumab-EU plus paclitaxel (trastuzumab-EU group; n = 355). PF-05280014 or trastuzumab-EU was administered weekly (first dose 4 mg/kg, subsequent doses 2 mg/kg), with the option to change to a 3-weekly regimen (6 mg/kg) from Week 33. Treatment with PF-05280014 or trastuzumab-EU could continue until disease progression. Paclitaxel (starting dose 80 mg/m
2 ) was administered on Days 1, 8 and 15 of 28-day cycles for at least six cycles or until maximal benefit of response. The primary endpoint was objective response rate (ORR), evaluating responses achieved by Week 25 and confirmed by Week 33, based on blinded central radiology review., Results: The risk ratio for ORR was 0.940 (95% CI: 0.842-1.049). The 95% CI fell within the pre-specified equivalence margin of 0.80-1.25. ORR was 62.5% (95% CI: 57.2-67.6%) in the PF-05280014 group and 66.5% (95% CI: 61.3-71.4%) in the trastuzumab-EU group. As of data cut-off on 11 January 2017 (using data up to 378 days post-randomisation), there were no notable differences between groups in progression-free survival (median: 12.16 months in the PF-05280014 group vs. 12.06 months in the trastuzumab-EU group; 1-year rate: 54% vs. 51%) or overall survival (median: not reached in either group; 1-year rate: 89.31% vs. 87.36%). Safety outcomes and immunogenicity were similar between the treatment groups., Conclusion: When given as first-line treatment for HER2-positive metastatic breast cancer, PF-05280014 plus paclitaxel demonstrated equivalence to trastuzumab-EU plus paclitaxel in terms of ORR., Clinical Trial Registration: ClinicalTrials.gov, NCT01989676.- Published
- 2019
- Full Text
- View/download PDF
32. Innovative Strategies: Targeting Subtypes in Metastatic Breast Cancer.
- Author
-
Pegram MD, Zong Y, Yam C, Goetz MP, and Moulder SL
- Subjects
- Antibody-Dependent Cell Cytotoxicity, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biomarkers, Tumor, Breast Neoplasms genetics, Breast Neoplasms metabolism, Combined Modality Therapy, Drug Resistance, Neoplasm genetics, Female, Humans, Immunomodulation, Molecular Targeted Therapy, Mutation, Neoplasm Metastasis, Neoplasm Staging, Polymorphism, Genetic, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Signal Transduction drug effects, Triple Negative Breast Neoplasms diagnosis, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms therapy, Breast Neoplasms diagnosis, Breast Neoplasms therapy
- Abstract
Metastatic breast cancer continues to be a life-threatening diagnosis that impacts hundreds of thousands of patients around the world. Targeted therapies are usually associated with less toxicity compared with cytotoxic chemotherapies and often induce response or durable disease control in estrogen receptor (ER) and/or HER2+ breast cancers. Drugs that target CDK 4/6 either alone or in combination with endocrine therapy have demonstrated substantial improvements in progression-free survival (PFS) compared with endocrine monotherapy. Most recently, PARP inhibitors have shown longer PFS compared with physician's choice of chemotherapy in BRCA-associated cancers, leading to the first U.S. Food and Drug Administration (FDA) approval of a targeted therapy with the potential to benefit a subgroup of patients with triple-negative breast cancer (TNBC). Finally, newer drug delivery strategies using antibody drug conjugates have also allowed a "targeted approach" to deliver moderate to extremely potent cytotoxins directly to sites of metastatic disease, with less toxicity.
- Published
- 2018
- Full Text
- View/download PDF
33. Anti-HER2 scFv-Directed Extracellular Vesicle-Mediated mRNA-Based Gene Delivery Inhibits Growth of HER2-Positive Human Breast Tumor Xenografts by Prodrug Activation.
- Author
-
Wang JH, Forterre AV, Zhao J, Frimannsson DO, Delcayre A, Antes TJ, Efron B, Jeffrey SS, Pegram MD, and Matin AC
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Female, Humans, MCF-7 Cells, Mice, Inbred BALB C, Mice, Nude, Oxazines metabolism, Prodrugs metabolism, RNA, Messenger genetics, Receptor, ErbB-2 immunology, Single-Chain Antibodies metabolism, Tumor Burden drug effects, Breast Neoplasms drug therapy, Extracellular Vesicles metabolism, RNA, Messenger metabolism, Receptor, ErbB-2 metabolism, Single-Chain Antibodies administration & dosage, Xenograft Model Antitumor Assays
- Abstract
This paper deals with specific targeting of the prodrug/enzyme regimen, CNOB/HChrR6, to treat a serious disease, namely HER2
+ human breast cancer with minimal off-target toxicity. HChrR6 is an improved bacterial enzyme that converts CNOB into the cytotoxic drug MCHB. Extracellular vesicles (EV) were used for mRNA-based H chrR6 gene delivery: EVs may cause minimal immune rejection, and mRNA may be superior to DNA for gene delivery. To confine HChrR6 generation and CNOB activation to the cancer, the EVHB chimeric protein was constructed. It contains high-affinity anti-HER2 scFv antibody (ML39) and is capable of latching on to EV surface. Cells transfected with EVHB-encoding plasmid generated EVs displaying this protein ("directed EVs"). Transfection of a separate batch of cells with the new plasmid, XPort/HChrR6, generated EVs containing HChrR6 mRNA; incubation with pure EVHB enabled these to target the HER2 receptor, generating "EXO-DEPT" EVs. EXO-DEPT treatment specifically enabled HER2-overexpressing BT474 cells to convert CNOB into MCHB in actinomycin D-independent manner, showing successful and specific delivery of HChrR6 mRNA. EXO-DEPTs-but not undirected EVs-plus CNOB caused near-complete growth arrest of orthotopic BT474 xenografts in vivo , demonstrating for the first time EV-mediated delivery of functional exogenous mRNA to tumors. EXO-DEPTs may be generated from patients' own dendritic cells to evade immune rejection, and without plasmids and their potentially harmful genetic material, raising the prospect of clinical use of this regimen. This approach can be used to treat any disease overexpressing a specific marker. Mol Cancer Ther; 17(5); 1133-42. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
34. Electrophoretic cytopathology resolves ERBB2 forms with single-cell resolution.
- Author
-
Kang CC, Ward TM, Bockhorn J, Schiffman C, Huang H, Pegram MD, and Herr AE
- Abstract
In addition to canonical oncoproteins, truncated isoforms and proteolysis products are implicated in both drug resistance and disease progression. In HER2-positive breast tumors, expression of truncated HER2 isoforms resulting from alternative translation and/or carboxy-terminal fragments (CTFs) resulting from proteolysis (collectively, t-erbB2) have been associated with shortened progression-free survival of patients. Thus, to advance clinical pathology and inform treatment decisions, we developed a high-selectivity cytopathology assay capable of distinguishing t-erbB2 from full-length HER2 expression without the need for isoform-specific antibodies. Our microfluidic, single-cell western blot, employs electrophoretic separations to resolve full-length HER2 from the smaller t-erbB2 in each ~28 pL single-cell lysate. Subsequently, a pan-HER2 antibody detects all resolved HER2 protein forms via immunoprobing. In analysis of eight breast tumor biopsies, we identified two tumors comprised of 15% and 40% t-erbB2-expressing cells. By single-cell western blotting of the t-erbB2-expressing cells, we observed statistically different ratios of t-erbB2 proteins to full-length HER2 expression. Further, target multiplexing and clustering analyses scrutinized signaling, including ribosomal S6, within the t-erbB2-expressing cell subpopulation. Taken together, cytometric assays that report both protein isoform profiles and signaling state offer cancer classification taxonomies with unique relevance to precisely describing drug resistance mechanisms in which oncoprotein isoforms/fragments are implicated., Competing Interests: C.-C.K., and A.E.H. are co-inventors on intellectual property related to the scWB device and assay described here and may benefit from royalties from licensing. A.E.H. has financial interest in commercialization efforts.
- Published
- 2018
- Full Text
- View/download PDF
35. Phase I study of the gamma secretase inhibitor PF-03084014 in combination with docetaxel in patients with advanced triple-negative breast cancer.
- Author
-
Locatelli MA, Aftimos P, Dees EC, LoRusso PM, Pegram MD, Awada A, Huang B, Cesari R, Jiang Y, Shaik MN, Kern KA, and Curigliano G
- Subjects
- Adult, Aged, Disease Progression, Docetaxel, Dose-Response Relationship, Drug, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neutropenia chemically induced, Neutropenia epidemiology, Taxoids adverse effects, Tetrahydronaphthalenes adverse effects, Triple Negative Breast Neoplasms epidemiology, Triple Negative Breast Neoplasms pathology, Valine administration & dosage, Valine adverse effects, Vomiting chemically induced, Vomiting epidemiology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Taxoids administration & dosage, Tetrahydronaphthalenes administration & dosage, Triple Negative Breast Neoplasms drug therapy, Valine analogs & derivatives
- Abstract
Background: The NOTCH signaling pathway may be involved in the survival of stem cell-like tumor-initiating cells and contribute to tumor growth. In this phase Ib, open-label, multicenter study (NCT01876251), we evaluated PF-03084014, a selective gamma-secretase inhibitor in patients with advanced triple-negative breast cancer., Methods: The dose-finding part was based on a 2×3 matrix design using the modified toxicity probability interval method. Oral PF-03084014 was administered twice daily continuously in combination with intravenous docetaxel given on day 1 of each 21-day cycle. Primary endpoint was first-cycle dose-limiting toxicity (DLT) for the dose-finding part and 6-month progression-free survival (PFS) for the expansion cohort treated at the maximum tolerated dose (MTD). Secondary endpoints included safety, objective response, and pharmacokinetics of the combination., Results and Conclusions: The MTD was estimated to be PF-03084014 100 mg twice daily / docetaxel 75 mg/m2. At this dose level, combination treatment was generally well tolerated (one DLT, grade 3 diarrhea, among eight DLT-evaluable patients). The most common all-grade, treatment-related adverse events reported in all patients (N = 29) were neutropenia (90%), fatigue (79%), nausea (72%), leukopenia (69%), diarrhea (59%), alopecia (55%), anemia (55%), and vomiting (48%). No effect was observed on the pharmacokinetics of docetaxel when administered in combination with PF-03084014. Four (16%) of 25 response-evaluable patients achieved a confirmed partial response; nine (36%) patients had stable disease, including five patients with unconfirmed partial response. In the expansion cohort, median PFS was 4.1 (95% CI 1.3-8.1) months (6-month PFS rate 17.1% [95% CI 0.8-52.6%]).
- Published
- 2017
- Full Text
- View/download PDF
36. Neratinib in ERBB2-Positive Brain Metastases.
- Author
-
Pegram MD
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Brain Neoplasms genetics, Brain Neoplasms pathology, Brain Neoplasms secondary, Breast Neoplasms genetics, Breast Neoplasms pathology, Female, Humans, Receptor, ErbB-2 genetics, Brain Neoplasms drug therapy, Breast Neoplasms drug therapy
- Published
- 2016
- Full Text
- View/download PDF
37. Relationship between Tumor Biomarkers and Efficacy in EMILIA, a Phase III Study of Trastuzumab Emtansine in HER2-Positive Metastatic Breast Cancer.
- Author
-
Baselga J, Lewis Phillips GD, Verma S, Ro J, Huober J, Guardino AE, Samant MK, Olsen S, de Haas SL, and Pegram MD
- Subjects
- Ado-Trastuzumab Emtansine, Animals, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Breast Neoplasms mortality, Breast Neoplasms pathology, Disease Models, Animal, Female, Follow-Up Studies, Humans, Kaplan-Meier Estimate, Maytansine administration & dosage, Maytansine adverse effects, Maytansine therapeutic use, Mice, Neoplasm Metastasis, Neoplasm Staging, Phosphatidylinositol 3-Kinases, Proportional Hazards Models, Retreatment, Trastuzumab administration & dosage, Trastuzumab adverse effects, Xenograft Model Antitumor Assays, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Maytansine analogs & derivatives, Receptor, ErbB-2 metabolism, Trastuzumab therapeutic use
- Abstract
Purpose: HER2-positive breast cancer is heterogeneous. Some tumors express mutations, like activating PIK3CA mutations or reduced PTEN expression, that negatively correlate with response to HER2-targeted therapies. In this exploratory analysis, we investigated whether the efficacy of trastuzumab emtansine (T-DM1), an antibody-drug conjugate comprised of the cytotoxic agent DM1 linked to the HER2-targeted antibody trastuzumab, was correlated with the expression of specific biomarkers in the phase III EMILIA study., Experimental Design: Tumors were evaluated for HER2 (n = 866), EGFR (n = 832), and HER3 (n = 860) mRNA expression by quantitative reverse transcriptase PCR; for PTEN protein expression (n = 271) by IHC; and for PIK3CA mutations (n = 259) using a mutation detection kit. Survival outcomes were analyzed by biomarker subgroups. T-DM1 was also tested on cell lines and in breast cancer xenograft models containing PIK3CA mutations., Results: Longer progression-free survival (PFS) and overall survival (OS) were observed with T-DM1 compared with capecitabine plus lapatinib in all biomarker subgroups. PIK3CA mutations were associated with shorter median PFS (mutant vs. wild type: 4.3 vs. 6.4 months) and OS (17.3 vs. 27.8 months) in capecitabine plus lapatinib-treated patients, but not in T-DM1-treated patients (PFS, 10.9 vs. 9.8 months; OS, not reached in mutant or wild type). T-DM1 showed potent activity in cell lines and xenograft models with PIK3CA mutations., Conclusions: Although other standard HER2-directed therapies are less effective in tumors with PI3KCA mutations, T-DM1 appears to be effective in both PI3KCA-mutated and wild-type tumors. Clin Cancer Res; 22(15); 3755-63. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
38. Classification of large circulating tumor cells isolated with ultra-high throughput microfluidic Vortex technology.
- Author
-
Che J, Yu V, Dhar M, Renier C, Matsumoto M, Heirich K, Garon EB, Goldman J, Rao J, Sledge GW, Pegram MD, Sheth S, Jeffrey SS, Kulkarni RP, Sollier E, and Di Carlo D
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms blood, Breast Neoplasms pathology, Female, Humans, Lung Neoplasms blood, Lung Neoplasms pathology, Male, Middle Aged, Neoplastic Cells, Circulating pathology, High-Throughput Screening Assays methods, Microfluidic Analytical Techniques methods, Neoplastic Cells, Circulating classification
- Abstract
Circulating tumor cells (CTCs) are emerging as rare but clinically significant non-invasive cellular biomarkers for cancer patient prognosis, treatment selection, and treatment monitoring. Current CTC isolation approaches, such as immunoaffinity, filtration, or size-based techniques, are often limited by throughput, purity, large output volumes, or inability to obtain viable cells for downstream analysis. For all technologies, traditional immunofluorescent staining alone has been employed to distinguish and confirm the presence of isolated CTCs among contaminating blood cells, although cells isolated by size may express vastly different phenotypes. Consequently, CTC definitions have been non-trivial, researcher-dependent, and evolving. Here we describe a complete set of objective criteria, leveraging well-established cytomorphological features of malignancy, by which we identify large CTCs. We apply the criteria to CTCs enriched from stage IV lung and breast cancer patient blood samples using the High Throughput Vortex Chip (Vortex HT), an improved microfluidic technology for the label-free, size-based enrichment and concentration of rare cells. We achieve improved capture efficiency (up to 83%), high speed of processing (8 mL/min of 10x diluted blood, or 800 μL/min of whole blood), and high purity (avg. background of 28.8±23.6 white blood cells per mL of whole blood). We show markedly improved performance of CTC capture (84% positive test rate) in comparison to previous Vortex designs and the current FDA-approved gold standard CellSearch assay. The results demonstrate the ability to quickly collect viable and pure populations of abnormal large circulating cells unbiased by molecular characteristics, which helps uncover further heterogeneity in these cells.
- Published
- 2016
- Full Text
- View/download PDF
39. Hydrogel Pore-Size Modulation for Enhanced Single-Cell Western Blotting.
- Author
-
Duncombe TA, Kang CC, Maity S, Ward TM, Pegram MD, Murthy N, and Herr AE
- Subjects
- Blotting, Western methods, Breast Neoplasms metabolism, Cell Line, Tumor, Electrophoresis, Polyacrylamide Gel instrumentation, Electrophoresis, Polyacrylamide Gel methods, Extracellular Signal-Regulated MAP Kinases metabolism, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Light, Nucleocytoplasmic Transport Proteins metabolism, Porosity, Receptor, ErbB-2 metabolism, Single-Cell Analysis methods, TOR Serine-Threonine Kinases metabolism, Blotting, Western instrumentation, Hydrogels chemistry, Hydrogels radiation effects, Single-Cell Analysis instrumentation
- Abstract
Pore-gradient microgel arrays enable thousands of parallel high-resolution single-cell protein electrophoresis separations for targets accross a wide molecular mass (25-289 kDa), yet within 1 mm separation distances. Dual crosslinked hydrogels facilitate gel-pore expansion after electrophoresis for efficient and uniform immunoprobing. The photopatterned, light-activated, and acid-expandable hydrogel underpins single-cell protein analysis, here for oncoprotein-related signaling in human breast biopsy., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
40. COUNTERPOINT: HER2-Targeted Combinations in Advanced HER2-Positive Breast Cancer.
- Author
-
Pegram MD
- Subjects
- Breast Neoplasms chemistry, Female, Humans, Receptor, ErbB-2 analysis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Receptor, ErbB-2 antagonists & inhibitors
- Published
- 2015
41. "Vertical" Inhibition of HER2 Yields Horizontal Gains in the Clinic.
- Author
-
Sledge GW and Pegram MD
- Subjects
- Female, Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Receptor, ErbB-2 metabolism
- Abstract
HER2-targeted therapy has moved beyond trastuzumab to include other monoclonals targeting the cell surface, receptor tyrosine kinase inhibitors of HER2, and antibody-drug conjugates. Afatinib, a small molecule receptor tyrosine kinase inhibitor, now joins the ranks of HER2-targeting agents in combination with trastuzumab. The combination brings new opportunities and challenges., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
42. Everything old is neu again: cellular senescence in HER2-positive breast cancer.
- Author
-
Sledge GW and Pegram MD
- Subjects
- Animals, Female, Humans, Biomarkers, Tumor analysis, Breast Neoplasms chemistry, Breast Neoplasms pathology, Cellular Senescence, Receptor, ErbB-2 analysis
- Published
- 2015
- Full Text
- View/download PDF
43. Personalization of loco-regional care for primary breast cancer patients (part 2).
- Author
-
Toi M, Winer EP, Benson JR, Inamoto T, Forbes JF, von Minckwitz G, Robertson JF, Grobmyer SR, Jatoi I, Sasano H, Kunkler I, Ho AY, Yamauchi C, Chow LW, Huang CS, Han W, Noguchi S, Pegram MD, Yamauchi H, Lee ES, Larionov AA, Bevilacqua JL, Yoshimura M, Sugie T, Yamauchi A, Krop IE, Noh DY, and Klimberg VS
- Subjects
- Disease Management, Female, Humans, Breast Neoplasms diagnosis, Breast Neoplasms therapy, Precision Medicine
- Abstract
Kyoto Breast Cancer Consensus Conference, Kyoto, Japan, 18-20 February 2014 The loco-regional management of breast cancer is increasingly complex with application of primary systemic therapies, oncoplastic techniques and genetic testing for breast cancer susceptibility. Personalization of loco-regional treatment is integral to optimization of breast cancer care. Clinical and pathological tumor stage, biological features and host factors influence loco-regional treatment strategies and extent of surgical procedures. Key issues including axillary staging, axillary treatment, radiation therapy, primary systemic therapy (PST), preoperative hormonal therapy and genetic predisposition were identified and discussed at the Kyoto Breast Cancer Consensus Conference (KBCCC2014). In the second of a two part conference scene, consensus recommendations for radiation treatment, primary systemic therapies and management of genetic predisposition are reported and focus on the following topics: influence of both clinical response to PST and stage at presentation on recommendations for postmastectomy radiotherapy; use of regional nodal irradiation in selected node-positive patients and those with adverse pathological factors; extent of surgical resection following downstaging of tumors with PST; use of preoperative hormonal therapy in premenopausal women with larger, node-negative luminal A-like tumors and managing increasing demands for contralateral prophylactic mastectomy in patients with a unilateral sporadic breast cancer.
- Published
- 2015
- Full Text
- View/download PDF
44. Personalization of loco-regional care for primary breast cancer patients (part 1).
- Author
-
Toi M, Winer EP, Benson JR, Inamoto T, Forbes JF, von Minckwitz G, Robertson JF, Grobmyer SR, Jatoi I, Sasano H, Kunkler I, Ho AY, Yamauchi C, Chow LW, Huang CS, Han W, Noguchi S, Pegram MD, Yamauchi H, Lee ES, Larionov AA, Bevilacqua JL, Yoshimura M, Sugie T, Yamauchi A, Krop IE, Noh DY, and Klimberg VS
- Subjects
- Disease Management, Female, Humans, Breast Neoplasms diagnosis, Breast Neoplasms therapy, Precision Medicine
- Abstract
ABSTRACT Kyoto Breast Cancer Consensus Conference, Kyoto, Japan, 18-20 February 2014 The loco-regional management of breast cancer is increasingly complex with application of primary systemic therapies, oncoplastic techniques and genetic testing for breast cancer susceptibility. Personalization of loco-regional treatment is integral to optimization of breast cancer care. Clinical and pathological tumor stage, biological features and host factors influence loco-regional treatment strategies and extent of surgical procedures. Key issues including axillary staging, axillary treatment, radiation therapy, primary systemic therapy (PST), preoperative hormonal therapy and genetic predisposition were identified and discussed at the Kyoto Breast Cancer Consensus Conference (KBCCC2014). In the first of a two part conference scene, consensus recommendations for axillary management are presented and focus on the following topics: indications for completion axillary lymph node dissection in primary surgical patients with ≤2 macrometastases or any sentinel nodal deposits after PST; the timing of sentinel lymph node biopsy in the context of PST; use of axillary irradiation as a component of primary treatment plans and the role of intraoperative node assessment in the post-Z0011 era.
- Published
- 2015
- Full Text
- View/download PDF
45. Rapid reduction in breast cancer mortality with inorganic arsenic in drinking water.
- Author
-
Smith AH, Marshall G, Yuan Y, Steinmaus C, Liaw J, Smith MT, Wood L, Heirich M, Fritzemeier RM, Pegram MD, and Ferreccio C
- Abstract
Background: Arsenic trioxide is effective in treating promyelocytic leukemia, and laboratory studies demonstrate that arsenic trioxide causes apoptosis of human breast cancer cells. Region II in northern Chile experienced very high concentrations of inorganic arsenic in drinking water, especially in the main city Antofagasta from 1958 until an arsenic removal plant was installed in 1970., Methods: We investigated breast cancer mortality from 1950 to 2010 among women in Region II compared to Region V, which had low arsenic water concentrations. We conducted studies on human breast cancer cell lines and compared arsenic exposure in Antofagasta with concentrations inducing apoptosis in laboratory studies., Findings: Before 1958, breast cancer mortality rates were similar, but in 1958-1970 the rates in Region II were half those in Region V (rate ratio RR = 0·51, 95% CI 0·40-0·66; p<0·0001). Women under the age of 60 experienced a 70% reduction in breast cancer mortality during 1965-1970 (RR=0·30, 0·17-0·54; p<0·0001). Breast cancer cell culture studies showed apoptosis at arsenic concentrations close to those estimated to have occurred in people in Region II., Interpretation: We found biologically plausible major reductions in breast cancer mortality during high exposure to inorganic arsenic in drinking water which could not be attributed to bias or confounding. We recommend clinical trial assessment of inorganic arsenic in the treatment of advanced breast cancer.
- Published
- 2014
- Full Text
- View/download PDF
46. Treating the HER2 pathway in early and advanced breast cancer.
- Author
-
Pegram MD
- Subjects
- Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms pathology, Female, Humans, Molecular Targeted Therapy, Neoplasm Staging, Receptor, ErbB-2 antagonists & inhibitors, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Receptor, ErbB-2 metabolism, Signal Transduction drug effects
- Abstract
ERBB2 gene amplification occurs in ∼20% of human breast cancers (BC) and is associated with an adverse clinical prognosis, indicating that it may be playing a critical role in disease pathogenesis. Therapeutic strategies targeting pathologic ERBB2 overexpression have revolutionized the diagnosis and treatment of BC. Indeed, humanized anti-ERBB2 antibodies, small molecule ERBB2 kinase inhibitors and ERBB2-targeting antibody-drug conjugates have proven safety and efficacy based upon evidence from randomized phase III clinical trials. Recent progress in targeting ERBB2 alteration will be reviewed, with focus on data that has informed changes in clinical practice for the treatment of BC., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Truncated p110 ERBB2 induces mammary epithelial cell migration, invasion and orthotopic xenograft formation, and is associated with loss of phosphorylated STAT5.
- Author
-
Ward TM, Iorns E, Liu X, Hoe N, Kim P, Singh S, Dean S, Jegg AM, Gallas M, Rodriguez C, Lippman M, Landgraf R, and Pegram MD
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Female, Humans, Immunohistochemistry, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Invasiveness, Peptide Fragments metabolism, Phosphorylation, STAT5 Transcription Factor genetics, Signal Transduction, Transplantation, Heterologous, Breast Neoplasms pathology, Cell Movement physiology, Receptor, ErbB-2 metabolism, STAT5 Transcription Factor metabolism
- Abstract
Truncated-ERBB2 isoforms (t-ERBB2s), resulting from receptor proteolysis or alternative translation of the ERBB2 mRNA, exist in a subset of human breast tumors. t-ERBB2s lack the receptor extracellular domain targeted by therapeutic anti-ERBB2 antibodies and antibody-drug conjugates, including trastuzumab, trastuzumab-DM1 and pertuzumab. In clinical studies, expression of t-ERBB2 in breast tumors correlates with metastasis as well as trastuzumab resistance. By using a novel immuno-microarray method, we detect a significant t-ERBB2 fraction in 18 of 31 (58%) of immunohistochemistry (IHC)3+ ERBB2+ human tumor specimens, and further show that t-ERBB2 isoforms are phosphorylated in a subset of IHC3+ samples (10 of 31, 32%). We investigated t-ERBB2 biological activity via engineered expression of full-length and truncated ERBB2 isoforms in human mammary epithelial cells (HMECs), including HMEC and MCF10A cells. Expression of p110 t-ERBB2, but not p95m (m=membrane, also 648CTF) or intracellular ERBB2s, significantly enhanced cell migration and invasion in multiple cell types. In addition, only expression of the p110 isoform led to human breast epithelial cell (HMLE) xenograft formation in vivo. Expression of t-ERBB2s did not result in hyperactivation of the phosphoinositide kinase-3/AKT or mitogen-activated protein kinase signaling pathways in these cells; rather, phosphoproteomic array profiling revealed attenuation of phosphorylated signal transducer and activator of transcription 5 (STAT5) in p110-t-ERBB2-expressing cells compared to controls. Short hairpin-mediated silencing of STAT5 phenocopied p110-t-ERBB2-driven cell migration and invasion, while expression of constitutively active STAT5 reversed these effects. Thus, we provide novel evidence that (1) expression of p110 t-ERBB2 is sufficient for full transformation of HMEC, yielding in vivo xenograft formation, and (2) truncated p110 t-ERBB2 expression is associated with decreased phosphorylation of STAT5.
- Published
- 2013
- Full Text
- View/download PDF
48. The use of neoadjuvant platinum-based chemotherapy in locally advanced breast cancer that is triple negative: retrospective analysis of 144 patients.
- Author
-
Hurley J, Reis IM, Rodgers SE, Gomez-Fernandez C, Wright J, Leone JP, Larrieu R, and Pegram MD
- Subjects
- Adult, Aged, Aged, 80 and over, Axilla pathology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carboplatin administration & dosage, Cisplatin administration & dosage, Disease-Free Survival, Docetaxel, Female, Humans, Middle Aged, Neoadjuvant Therapy, Retrospective Studies, Taxoids administration & dosage, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms mortality
- Abstract
Triple-negative breast cancers comprise about 20 % of breast cancers. They have poor prognosis and have no standard therapy. The aim of this study was to evaluate pathologic complete response (pCR), progression-free survival (PFS), and overall survival (OS) in patients with TNBC treated with neoadjuvant platinum-based chemotherapy. This is a retrospective study of one hundred and forty-four women with TNBC treated with neoadjuvant platinum-containing chemotherapy for locally advanced breast cancer at the University of Miami between January 1, 1999, and January 1, 2011. The medical record was reviewed to obtain data on clinical characteristics, including ethnicity, race, age, clinical stage, treatment regimen, and vital status. This study was approved by the University of Miami IRB. All patients had locally advanced breast cancer with at least one of the following features at presentation: T3, T4, N2, and N3. The mean tumor size by palpation was 9.4 cm. The clinical T-stage at presentation was 1.4 % T1, 8.3 % T2, 52.8 % T3, and 37.5 % T4 (19.4 % T4d). The nodal status by physical exam at presentation was 23 % N0, 37.5 % N1, 34 % N2, and 5.5 % N3. pCR in breast and axilla was seen in 31 %. PFS and OS were 55 and 59 %, respectively, at 7 years. Cisplatin offered a survival advantage over carboplatin in both PFS (P = 0.007) and OS (P = 0.018). Node positivity was the most important predictor of survival. Cisplatin/docetaxel neoadjuvant therapy was well tolerated and an effective therapy in locally advanced TNB.
- Published
- 2013
- Full Text
- View/download PDF
49. PI3K independent activation of mTORC1 as a target in lapatinib-resistant ERBB2+ breast cancer cells.
- Author
-
Jegg AM, Ward TM, Iorns E, Hoe N, Zhou J, Liu X, Singh S, Landgraf R, and Pegram MD
- Subjects
- Breast Neoplasms metabolism, Cell Line, Tumor, Female, Humans, Lapatinib, Mechanistic Target of Rapamycin Complex 1, Multiprotein Complexes, Mutation, Oncogene Protein v-akt metabolism, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Phosphatidylinositol 3-Kinases genetics, Phosphorylation drug effects, Receptor, ErbB-2 genetics, TOR Serine-Threonine Kinases, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Phosphatidylinositol 3-Kinases metabolism, Proteins metabolism, Quinazolines pharmacology, Receptor, ErbB-2 metabolism
- Abstract
Therapies targeting the ERBB2 receptor, including the kinase inhibitor lapatinib (Tykerb, GlaxoSmithKline), have improved clinical outcome for women with ERBB2-amplified breast cancer. However, acquired resistance to lapatinib remains a significant clinical problem, and the mechanisms governing resistance remain poorly understood. We sought to define molecular alterations that confer an acquired lapatinib resistance phenotype in ER-/ERBB2+ human breast cancer cells. ERBB2-amplified SKBR3 breast cancer cells were rendered resistant to lapatinib via culture in increasing concentrations of the drug, and molecular changes associated with a resistant phenotype were interrogated using a collaborative enzyme-enhanced immunoassay platform and immunoblotting techniques for detection of phosphorylated signaling cascade proteins. Interestingly, despite apparent inactivation of the PI3K/AKT signaling pathway, resistant cells exhibited constitutive activation of mammalian target of rapamycin complex 1 (mTORC1) and were highly sensitive to mTOR inhibition with rapamycin and the dual PI3K/mTOR inhibitor NVP-BEZ235. These data demonstrate a role for downstream activation of mTORC1 in the absence of molecular alterations leading to PI3K/AKT hyperactivation as a potential mechanism of lapatinib resistance in this model of ERBB2+ breast cancer and support the rationale of combination or sequential therapy using ERBB2 and mTOR-targeting molecules to prevent or target resistance to lapatinib. Moreover, our data suggest that assessment of mTOR substrate phosphorylation (i.e., S6) may serve as a more robust biomarker to predict sensitivity to mTOR inhibitors in the context of lapatinib resistance than PI3K mutations, loss of PTEN and p-AKT levels.
- Published
- 2012
- Full Text
- View/download PDF
50. Preoperative systemic therapy in locoregional management of early breast cancer: highlights from the Kyoto Breast Cancer Consensus Conference.
- Author
-
Toi M, Benson JR, Winer EP, Forbes JF, von Minckwitz G, Golshan M, Robertson JF, Sasano H, Cole BF, Chow LW, Pegram MD, Han W, Huang CS, Ikeda T, Kanao S, Lee ES, Noguchi S, Ohno S, Partridge AH, Rouzier R, Tozaki M, Sugie T, Yamauchi A, and Inamoto T
- Subjects
- Algorithms, Axilla surgery, Breast Neoplasms metabolism, Female, Humans, Lymphatic Metastasis pathology, Neoadjuvant Therapy, Nomograms, Sentinel Lymph Node Biopsy, Tumor Burden, Breast Neoplasms pathology, Breast Neoplasms surgery, Breast Neoplasms therapy, Preoperative Care
- Abstract
Data reviewed at the Kyoto Breast Cancer Consensus Conference (KBCCC) showed that preoperative systemic therapy (PST) could optimize surgery through the utilization of information relating to pre- and post-PST tumor stage, therapeutic sensitivity, and treatment-induced changes in the biological characteristics of the tumor. As such, it was noted that the biological characteristics of the tumor, such as hormone receptors, human epidermal growth factor receptor-2, histological grade, cell proliferative activity, mainly defined by the Ki67 labeling index, and the tumor's multi-gene signature, should be considered in the planning of both systemic and local therapy. Furthermore, the timing of axillary sentinel lymph node diagnosis (i.e., before or after the PST) was also noted to be critical in that it may influence the likelihood of axillary preservation, even in node positive cases. In addition, axillary diagnosis with ultrasound and concomitant fine needle aspiration cytology or core needle biopsy (CNB) was reported to contribute to the construction of a treatment algorithm for patient-specific or individualized axillary surgery. Following PST, planning for breast surgery should therefore be based on tumor subtype, tumor volume and extent, therapeutic response to PST, and patient preference. Nomograms for predicting nodal status and drug sensitivity were also recognized as a tool to support decision-making in the selection of surgical treatment. Overall, review of data at the KBCCC showed that PST increases the likelihood of patients receiving localized surgery and individualized treatment regimens.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.