Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael J. Berger is active.

Publication


Featured researches published by Michael J. Berger.


Breast Cancer Research | 2014

Heterogeneous atypical cell populations are present in blood of metastatic breast cancer patients

Maryam B. Lustberg; Priya Balasubramanian; Brandon A. Miller; Alejandra Garcia-Villa; Clayton Deighan; Yongqi Wu; Sarah Carothers; Michael J. Berger; Bhuvaneswari Ramaswamy; Erin Macrae; Robert Wesolowski; Rachel Layman; Ewa Mrozek; Xueliang Pan; Thomas A. Summers; Charles L. Shapiro; Jeffrey J. Chalmers

IntroductionCirculating tumor cells (CTCs) are commonly isolated from the blood by targeting the epithelial cell adhesion molecule (EpCAM) through positive selection. However, EpCAM can be downregulated during metastatic progression, or it can be initially not present. We designed the present prospective trial to characterize CTCs as well as other circulating cell populations in blood samples from women with metastatic breast cancer without EpCAM-dependent enrichment and/or isolation technology.MethodsA total of 32 patients with metastatic breast cancer were enrolled, and blood samples were processed using a previously described negative depletion immunomagnetic methodology. Samples from healthy volunteers were run as controls (n = 5). Multistep sequential labeling was performed to label and fix cell-surface markers followed by permeabilization for cytokeratins (CK) 8, 18 and 19. Multiparametric flow cytometry (FCM) analysis was conducted using a BD LSR II flow cytometer or a BD FACSAria II or FACSAria III cell sorter. Immunocytochemical staining on postenrichment specimens for DAPI, EpCAM, CD45, CK, epidermal growth factor receptor and vimentin was performed. Expression of these markers was visualized using confocal microscopy (CM).ResultsCD45-negative/CK-positive (CD45− CK+) populations with EpCAM + and EpCAM − expression were identified with both FCM and CM from the negatively enriched patient samples. In addition, EpCAM + and EpCAM − populations that were CK + and coexpressing the pan-hematopoietic marker CD45 were also noted. There were more CK + EpCAM − events/ml than CK + EpCAM + events/ml in both the CD45− and CD45+ fractions (both statistically significant at P ≤ 0.0005). The number of CK + CD45− and CK + CD45+ events per milliliter in blood samples (regardless of EpCAM status) was higher in patient samples than in normal control samples (P ≤ 0.0005 and P ≤ 0.026, respectively). Further, a significant fraction of the CK + CD45+ events also expressed CD68, a marker associated with tumor-associated macrophages. Higher levels of CD45-CK + EpCAM − were associated with worse overall survival (P = 0.0292).ConclusionsMetastatic breast cancer patients have atypical cells that are CK + EpCAM − circulating in their blood. Because a substantial number of these patients do not have EpCAM + CTCs, additional studies are needed to evaluate the role of EpCAM − circulating cells as a prognostic and predictive marker.


Journal of Clinical Oncology | 2016

T-DM1 Activity in Metastatic Human Epidermal Growth Factor Receptor 2–Positive Breast Cancers That Received Prior Therapy With Trastuzumab and Pertuzumab

Hannah Dzimitrowicz; Michael J. Berger; Craig Vargo; Annette Hood; Osama Abdelghany; Akshara Raghavendra; Debu Tripathy; Vicente Valero; Christos Hatzis; Lajos Pusztai; Rashmi Krishna Murthy

Purpose Ado-trastuzumab emtansine (T-DM1) is currently approved for treatment in patients with human epidermal growth factor receptor 2 (HER2)-positive, metastatic breast cancer (MBC) who previously received trastuzumab and a taxane. However, there are no data on the activity of T-DM1 in patients who received prior pertuzumab, which is now included as standard first-line therapy. The goal of this study was to assess the efficacy of T-DM1 in routine clinical practice in a contemporary patient population that received both prior trastuzumab and pertuzumab. Patients and Methods We identified all patients with HER2-positive MBC who received T-DM1 after trastuzumab and pertuzumab between March 1, 2013, and July 15, 2015, via electronic pharmacy records and departmental databases at three institutions: MD Anderson Cancer Center, Smilow Cancer Hospital at Yale, and The James Cancer Hospital at the Ohio State University. We reviewed medical records of each case to confirm treatment sequencing and outcome. Results Of patients, 82 were identified and 78 were available for outcome analysis; 32% received T-DM1 as first- and second-line line therapy, and 48% received it as fourth-line treatment or later. Rate of prolonged duration on therapy, defined as duration on therapy ≥ 6 months, was 30.8% (95% CI, 20.6% to 41.1%), and tumor response rate was 17.9% (95% CI, 9.4% to 26.4%). Median duration on therapy was 4.0 months (95% CI, 2.7 to 5.1; range, 0 to 22.5 months). T-DM1 was discontinued for disease progression in 84% of patients and for toxicity in 10%. Conclusion Tumor response rates were lower than in prior reports of trastuzumab-resistant, HER2-positive MBC, but one third of patients received therapy with T-DM1 for ≥ 6 months, which suggests a clinically relevant benefit in patients who received prior pertuzumab.


Journal of The National Comprehensive Cancer Network | 2017

Antiemesis, version 2.2017 featured updates to the NCCN guidelines

Michael J. Berger; David S. Ettinger; Jonathan Aston; Sally Barbour; Jason Bergsbaken; Philip J. Bierman; Debra S. Brandt; Dawn E. Dolan; Georgiana K. Ellis; Eun Jeong Kim; Steve Kirkegaard; Dwight D. Kloth; Ruth Lagman; Dean Lim; Charles L. Loprinzi; Cynthia X. Ma; Victoria Maurer; Laura Boehnke Michaud; Lisle Nabell; Kim Noonan; Eric Roeland; Hope S. Rugo; Lee S. Schwartzberg; Bridget Scullion; John Timoney; Barbara Todaro; Susan G. Urba; Dorothy A. Shead; Miranda Hughes

The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Antiemesis address all aspects of management for chemotherapy-induced nausea and vomiting. These NCCN Guidelines Insights focus on recent updates to the NCCN Guidelines for Antiemesis, specifically those regarding carboplatin, granisetron, and olanzapine.


Cancer Treatment Communications | 2015

Stabilization of bone marrow infiltration by metastatic breast cancer with continuous doxorubicin

Gaurav Pahouja; Robert Wesolowski; Raquel E. Reinbolt; Gary Tozbikian; Michael J. Berger; Neha Mangini; Maryam B. Lustberg

Complete bone marrow infiltration with profound pancytopenia is very uncommon in breast cancer. Bone marrow metastasis can frequently occur following development of metastatic breast cancer. However, bone marrow failure as the herald of this disease is not typically seen. Very limited data exists as to the safest and most efficacious manner to treat patients with profound pancytopenia due to metastatic solid tumor involvement. In this case, the patient’s thrombocytopenia was particularly worrisome, requiring daily platelet transfusions. There was also concern that cytotoxic chemotherapy would exacerbate the patient’s thrombocytopenia and increase bleeding risk. The patient’s dramatic response to chemotherapy with full platelet recovery is also highly unusual. For our patient, continuous doxorubicin successfully “unpacked” the bone marrow despite a low baseline platelet level, and without increasing the need for more frequent platelet transfusion or risk of catastrophic bleeding. Given the rarity of this presentation, it is currently unknown if the majority of similar patients experience near full recovery of hematopoietic function after initiation of appropriate systemic treatment for metastatic disease.


Journal of Clinical Oncology | 2012

Circulating CD68 positive (+) leukocytes in blood samples from patients (pts) with breast cancer (BC).

Maryam B. Lustberg; Brandon L. Miller; Yongqi Wu; Wei Xue; Clayton Deighan; Sarah Carothers; Michael J. Berger; Charles L. Shapiro; Jeffrey J. Chalmers

172 Background: Several different CD45 + leukocyte populations are present in primary breast tumors and stroma (Coussens et al, 2011). These include CD4+/CD8+ T cells and myeloid derived cells such tumor associated macrophages/monocytes (TAMs). TAMs are under investigation as prognostic and predictive markers during chemotherapy (CX). The detection of TAMs has been restricted thus far to tumor tissue. We hypothesized that we can detect changes in CD45+ leukocyte populations in blood samples obtained before and during CX from pts with early and advanced BC. METHODS Venous blood samples from BC patients were collected, and were either directly analyzed or further processed using previously described immunomagnetic negative depletion. Multistep, sequential labeling was performed to first label and fix cell surface markers followed by permeablization for cytokeratins, before and after negative depletion, followed by multiparameter flow cytometry analysis for the following basic markers: CD45, cytokeratins, and EpCAM, and for a subset of patients additional markers including: CD13, CD14, CD68, and CD133. RESULTS Forty blood samples were analyzed and study is ongoing. Different CD45+ subpopulations were observed in peripheral blood including: a) CD45+, CK+, CD68 negative (-), b) CD45+, CK+, CD68+, c) CD45+, CK+, CD68+, CD14+, and CD16+. In addition, there was a trend in increasing CD 68+ leukocytes after 1 cycle of CX in pts with poor response to therapy or progression of disease. No abnormal cell subpopulations were present blood samples from healthy volunteers. CONCLUSIONS This is the first study to report various peripheral blood leukocyte populations in BC that are similar to those observed in primary breast tumor stroma. The precise origin of these CD45 + cells is under investigation, including additional phenotypic characterization for TAMs. Results will be correlated to patient stage, tumor subtype and response to therapy.


Journal of Oncology Pharmacy Practice | 2018

Time to treatment failure of palbociclib and letrozole as second-line therapy or beyond in hormone receptor-positive advanced breast cancer

M Alexandra Schickli; Michael J. Berger; Maryam B. Lustberg; Marilly Palettas; Craig Vargo

Purpose The management of endocrine therapy resistance is one of the most challenging facets of advanced breast cancer treatment. Palbociclib is an inhibitor of cyclin-dependent kinases 4 and 6 approved for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer in combination with fulvestrant in postmenopausal women with disease progression following endocrine therapy. However, treatment responsiveness of tumors to palbociclib after multiple lines of endocrine therapy is not clearly established. The purpose of this study was to determine the efficacy of palbociclib and letrozole in patients pretreated with one or more lines of endocrine therapy. Methods This was a single-center, retrospective cohort study of all postmenopausal hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer patients who received palbociclib and letrozole as a second-line endocrine therapy or beyond (and no prior cyclin-dependent kinases 4 and 6 inhibitor therapy) between February 1, 2015, and July 31, 2016. The primary objective was to evaluate time to treatment failure of palbociclib in combination with letrozole as a second-line of therapy or beyond. Results Fifty-three patients meeting eligibility criteria were included in the analysis. For the primary outcome, the median time to treatment failure of palbociclib and letrozole was 6.3 months (95% CI 3.1–7.4 months). Progression-free survival of palbociclib and letrozole therapy was 6.4 months (95% CI 4.9–8.3 months). Conclusions Palbociclib and letrozole therapy is a viable, effective treatment option for metastatic breast cancer patients who were not exposed to cyclin-dependent kinases 4 and 6 inhibitors as a first-line endocrine therapy. The benefits of palbociclib and letrozole therapy were seen without excessive toxicity, and although neutropenia was common, it may be managed with dose reduction.


Journal of Oncology Pharmacy Practice | 2018

Updated report on incidence of infusion-site reactions associated with peripheral intravenous administration of fosaprepitant

Eric Chau; Jordan Lundberg; Gary Phillips; Michael J. Berger; Robert Wesolowski

Purpose Fosaprepitant (Emend®) is an antiemetic frequently used for the prevention of chemotherapy-induced nausea and vomiting. We previously documented an overall 28.7% incidence of infusion-site reactions in patients receiving fosaprepitant via peripheral venous access. These data resulted in a practice change within our institution; fosaprepitant is administered in more dilute concentrations over 30 min to prevent these adverse events. This retrospective study explored the impact of this practice change on the incidence of infusion-site reactions. Methods Medical records of patients with cancer receiving intravenous fosaprepitant through a peripheral intravenous line were reviewed. The primary objective of this study was to compare the incidence of infusion-site reactions before the practice change to the incidence after the practice change. Data collection included demographics, fosaprepitant infusion information, and grading of reactions. Results Between September 2013 and December of 2013, charts of 122 patients receiving intravenous fosaprepitant through a peripheral line at the The Arthur G. James Cancer Hospital at The Ohio State University were reviewed. We found a 5.74% incidence of infusion-site reactions which is significantly lower than the prechange incidence of 28.7% (p < 0.001). Conclusions Infusion-site reactions were significantly reduced when fosaprepitant was diluted to 150 mg/250 ml and infused over 30 min. We recommend oncology pharmacists consider using the more dilute fosaprepitant preparation and 30 min infusion duration when administering via a peripheral intravenous line to improve patient tolerance.


Cancer Research | 2018

Abstract OT2-05-03: Does minocycline mitigate chemotherapy induced neuroinflammation? A phase II randomized placebo controlled study

H Boutrid; Raquel E. Reinbolt; Michael V. Knopp; N Williams; J VanDeusen; S Sardesai; Anne M. Noonan; L Flora; E Gleich; X Pan; Michael J. Berger; C Vargo; Robert Wesolowski; Bhuvaneswari Ramaswamy; Ac DeVries; Maryam B. Lustberg

Background: Many breast cancer (BC) patients, particularly those who receive chemotherapy (chemo), experience affective symptoms and cognitive changes that can negatively impact their quality of life. Causal links between inflammatory mediators and the development of depressive-like behavior and cognitive defects, have been established in mouse models, including studies by our group showing increased microglial activation following chemo (A.C DeVries et al). Microglia are resident immune cells of the brain, which release proinflammatory cytokines when activated. Doxorubicin (DOX) induces microglial activation in the brain. Minocycline, a second generation tetracycline, has been shown to suppress inflammation by inhibiting microglial activation in CNS disease models. We hypothesize that (1) chemo activates microglia in the brains of women being treated for BC, which can precipitate or exacerbate depression, anxiety and cognitive deficits and (2) Minocycline administration during neoadjuvant or adjuvant chemo will prevent chemo-induced microglial activation and will reduce affective and cognitive symptom burden. Trial Design: This is a single center, Phase II, double blinded randomized study of minocycline (100 mg twice a day) vs placebo twice a day in women with BC receiving DOX-based or other chemo for BC. Pts will be randomized to either oral minocycline or placebo for up to a 1 week loading period plus chemo treatment period and an optional subsequent 2 week period. Eligibility Criteria: Women diagnosed with BC stages I-III initiating first line adjuvant or neoadjuvant chemo. Aims: (1) to evaluate symptoms related to anxiety and depression and cognitive changes during and after chemo completion (2) to evaluate markers of neuro inflammation as assessed by blood based inflammatory cytokines and central markers of inflammation and microglia activation using 1 F-Fludeoxyglucose and 11C-PK11195 positron emission tomography. Primary endpoints are changes in Center for Epidemiological Studies Depression Scale (CES-D) and State Trait Anxiety Index (STAI) from baseline to end of study after minocycline vs placebo intervention. Secondary endpoints are changes in cognitive function during chemo using validated cognitive testing including N-Back Test, Behavioural Rating Inventory of Executive Function (BRIEF) and the Multifactorial Memory Questionnaire Ability Scale (MMQ). Statistical Methods: Primary analysis for efficacy will be intention-to-treat. The main objective is to preliminarily evaluate the effect of minocycline on chemo-induced depressive symptoms in terms of changes in CES-D and STAI scores. Mixed models will be used to evaluate cognitive function changes. A sample size of 23 per group, will give 80% power to detect an effect size of 0.74 standard deviation (SD) difference between the 2 groups at significance level of 0.10 based on a 2 sided two-sample t-test. From our experience, attrition of less than 20% is expected for studies in this patient population in our center, and to account for this, we plan to recruit up to 60 patients. 16 of 46 evaluable pts have been accrued to date. Accrual started in January 2016. Funded by Pelotonia grant from The OSUCCC. Contact: Study PI: [email protected] Citation Format: Boutrid H, Reinbolt R, Knopp M, Williams N, VanDeusen J, Sardesai S, Noonan A, Flora L, Gleich E, Pan X, Berger M, Vargo C, Wesolowski R, Ramaswamy B, DeVries AC, Lustberg M. Does minocycline mitigate chemotherapy induced neuroinflammation? A phase II randomized placebo controlled study [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr OT2-05-03.


Cancer Research | 2015

Abstract P1-09-03: Prevention of aromatase Inhibitor (AI)-induced joint symptoms with omega-3 fatty acid supplementation: A randomized placebo-controlled pilot study

Maryam B. Lustberg; Tonya Orchard; Xueliang Pan; Raquel E. Reinbolt; Amanda Logan; Joanne Lester; Rachel Layman; Erin Macrae; Ewa Mrozek; Bhuvaneswari Ramaswamy; Robert Wesolowski; Michael J. Berger; Michael V. Knopp; Charles L. Loprinzi; Charles L. Shapiro; Lisa Yee

Introduction: AI-induced joint symptoms negatively impact drug adherence and quality of life. Based on observations that n-3 polyunsaturated fatty acids (PUFAs) have anti-inflammatory effects and that the mechanism of AI-induced joint symptoms may be partly due to inflammation, we hypothesized that women taking more n-3 PUFAs are less likely to develop AI-induced joint symptoms. Methods: We conducted a randomized, double-blind, placebo-controlled study comparing n-3 PUFA vs placebo in postmenopausal breast cancer patients starting adjuvant AIs. Participants were randomized to n-3 supplements [2.58 g eicosapentaenoic acid + 1.74 g docosahexaenoic acid/day; Marine Nutriceuticals, Mt. Bethel, PA] vs matched placebo for 24 weeks (wks). Primary endpoints was feasibility; secondary outcomes were self-reported symptoms as assessed by the Brief Pain Inventory short form (BPI-SF), Functional Assessment of Cancer Treatment, Breast & Endocrine Symptoms (FACTB-ES), and Stanford9s Health Assessment and Disability Index (HAQ) at baseline prior to AI receipt, 12 and 24 wks. Compliance and toxicity were evaluated monthly. Serial peripheral blood n-3 PUFA levels and inflammatory cytokines (IL-6, TNFR2, IL-17) were drawn. MRI of hands/wrists was performed in selected patients using a 3 Tesla dedicated wrist coil at baseline and treatment end. Results: Forty-four women were enrolled and randomized to study drug; 42 received ≥1 cycle (4 wks) of treatment; 36 had ≥1 post treatment evaluation at wk 12 or 24. Median age was 59.5 (range 43-76); history of prior taxane (n=15, 34%). The two groups’ baseline characteristics were similar. Overall, 93% and 88% of patients took >80% of the placebo and n-3 PUFA doses, respectively. Baseline erythrocyte n-3 PUFA was similar for both groups (6.6% ± 1.6%, 7.2% ±1.9%, p=0.20), but higher in the n-3 PUFA arm by wk 24 (6.5%±1.0% vs 15.0%±3.3%, p Conclusions: This is the first randomized pilot study to show that n-3 PUFA supplementation to prevent AI-induced joint symptoms is feasible and well tolerated. There is preliminary evidence that this intervention may help reduce the burden of AI-induced arthralgias. OSU Study #11022; ClinicalTrials.gov Identifier: NCT01478477. Grants from the National Cancer Institute (CA037447-26) to the Alliance for Clinical Trials in Oncology supported this pilot study. Citation Format: Maryam B Lustberg, Tonya Orchard, Xueliang Pan, Raquel Reinbolt, Amanda Logan, Joanne Lester, Rachel M Layman, Erin Macrae, Ewa Mrozek, Bhuvaneswari Ramaswamy, Robert Wesolowski, Michael Berger, Michael Knopp, Charles Loprinzi, Charles L Shapiro, Lisa Yee. Prevention of aromatase Inhibitor (AI)-induced joint symptoms with omega-3 fatty acid supplementation: A randomized placebo-controlled pilot study [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P1-09-03.


Cancer Research | 2010

Abstract 3284: Isolation of circulating tumor cells (CTCs) with mesenchymal and stem cell markers in localized and metastatic breast cancer using a novel negative selection enrichment

Maryam B. Lustberg; Priya Balasubramanian; Jas C. Lang; Amy S. Ruppert; Sarah Carothers; Michael J. Berger; Ewa Mrozek; Bhuvaneswari Ramaswamy; Rachel C. Layman; Jeffrey J. Chalmers; Charles L. Shapiro

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Background: Breast circulating tumor cells (CTCs) are commonly isolated by positive selection enrichment technology, which targets the epithelial cell adhesion activating molecule, EpCAM. However, CTCs with low or no EpCAM expression, such as those from basal and normal-like subtypes, are more likely to be missed by this method. We developed a novel negative enrichment technology to detect CTCs with mesenchymal and stem cell markers in localized and metastatic breast cancer (BC). Patients and Methods: Twenty nine patients with localized and metastatic BC initiating chemotherapy were enrolled. CTCs were isolated in 10 mL of peripheral blood using negative selection with immunomagnetic tagging and removal of CD45 positive cells at 3 time points: pretreatment, after one cycle of treatment (TP1) and at end of treatment or at disease progression in metastatic patients (TP2). Immunocytochemical staining for nucleus, cytokeratin (8,18,19), vimentin, and CD44 was completed on available samples. Double staining for nucleus and cytokeratin with high nucleus to cytoplasm ratio defined a CTC. Enrollment is ongoing. Results: Median age was 57 yrs (range 28-78 yrs); stage distributions were I-2 (7%), II-10 (35%), III-3 (10%), and IV-14 (48%); 19 (66%) were estrogen receptor positive (ER+), 4 (14%) estrogen and progesterone receptor negative (ER-PR-HER2 non-overexpressing) and 6 (21%) HER2 overexpressing. Negative enrichment yielded an average log10 depletion of nucleated cells of 2.74 and an overall, average log10 depletion of 5.2 (>100,000 enrichment). No CTCs were identified in 5 healthy volunteers or in buffy coats purchased from the Red Cross. CTCs were identified in all stages at pretreatment but decreased to 0 in three patients by TP1. Baseline median CTC level was 373/mL in localized BC (range 2.2-1975/mL) and 761/mL in metastatic BC (range 9.9-47513/mL). In localized BC, the median percent (%) change in CTCs was +2% at TP1 (n= 13; range −100% to +14945%) and −95% at TP2 (n=5; range −100% to −13%). In metastatic BC, median CTC numbers decreased by 41% at TP1 (n=11; range −100% to +4222%) and increased at TP2 in 2 patients by 231% and 730%. Baseline CTC levels and changes at TP1 were not significantly different between localized and metastatic groups by the Wilcoxen Rank Sum test. All tested CTCs expressed vimentin and CD44. In addition to CTCs, a population of cells without detectable cytokeratin expression but positive for the other markers was identified in some samples. Further characterization of these cells for epithelial mesenchymal transition markers is underway. Conclusions: CTCs identified with this novel negative selection method have both mesenchymal and stem cell markers in localized and metastatic BC. Higher CTC numbers with epithelial characteristics are detected with this method relative to what is reported with positive selection. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3284.

Collaboration


Dive into the Michael J. Berger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles L. Shapiro

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barbara Todaro

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge