Network


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

Hotspot


Dive into the research topics where Yvonne A. Efebera is active.

Publication


Featured researches published by Yvonne A. Efebera.


Cancer Cell | 2014

Widespread Genetic Heterogeneity in Multiple Myeloma: Implications for Targeted Therapy

Jens Lohr; Petar Stojanov; Scott L. Carter; Peter Cruz-Gordillo; Michael S. Lawrence; Daniel Auclair; Carrie Sougnez; Birgit Knoechel; Joshua Gould; Gordon Saksena; Kristian Cibulskis; Aaron McKenna; Michael Chapman; Ravid Straussman; Joan Levy; Louise M. Perkins; Jonathan J. Keats; Steven E. Schumacher; Mara Rosenberg; Kenneth C. Anderson; Paul G. Richardson; Amrita Krishnan; Sagar Lonial; Jonathan L. Kaufman; David Siegel; David H. Vesole; Vivek Roy; Candido E. Rivera; S. Vincent Rajkumar; Shaji Kumar

We performed massively parallel sequencing of paired tumor/normal samples from 203 multiple myeloma (MM) patients and identified significantly mutated genes and copy number alterations and discovered putative tumor suppressor genes by determining homozygous deletions and loss of heterozygosity. We observed frequent mutations in KRAS (particularly in previously treated patients), NRAS, BRAF, FAM46C, TP53, and DIS3 (particularly in nonhyperdiploid MM). Mutations were often present in subclonal populations, and multiple mutations within the same pathway (e.g., KRAS, NRAS, and BRAF) were observed in the same patient. In vitro modeling predicts only partial treatment efficacy of targeting subclonal mutations, and even growth promotion of nonmutated subclones in some cases. These results emphasize the importance of heterogeneity analysis for treatment decisions.


Blood | 2010

The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody.

Don M. Benson; Courtney E. Bakan; Anjali Mishra; Craig C. Hofmeister; Yvonne A. Efebera; Brian Becknell; Robert A. Baiocchi; Jianying Zhang; Jianhua Yu; Megan K Smith; Carli N Greenfield; Pierluigi Porcu; Steven M. Devine; Rinat Rotem-Yehudar; Gerard Lozanski; John C. Byrd; Michael A. Caligiuri

T-cell expression of programmed death receptor-1 (PD-1) down-regulates the immune response against malignancy by interacting with cognate ligands (eg, PD-L1) on tumor cells; however, little is known regarding PD-1 and natural killer (NK) cells. NK cells exert cytotoxicity against multiple myeloma (MM), an effect enhanced through novel therapies. We show that NK cells from MM patients express PD-1 whereas normal NK cells do not and confirm PD-L1 on primary MM cells. Engagement of PD-1 with PD-L1 should down-modulate the NK-cell versus MM effect. We demonstrate that CT-011, a novel anti-PD-1 antibody, enhances human NK-cell function against autologous, primary MM cells, seemingly through effects on NK-cell trafficking, immune complex formation with MM cells, and cytotoxicity specifically toward PD-L1(+) MM tumor cells but not normal cells. We show that lenalidomide down-regulates PD-L1 on primary MM cells and may augment CT-011s enhancement of NK-cell function against MM. We demonstrate a role for the PD-1/PD-L1 signaling axis in the NK-cell immune response against MM and a role for CT-011 in enhancing the NK-cell versus MM effect. A phase 2 clinical trial of CT-011 in combination with lenalidomide for patients with MM should be considered.


Blood | 2012

A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma.

Don M. Benson; Craig C. Hofmeister; Swaminathan Padmanabhan; Attaya Suvannasankha; Sundar Jagannath; Rafat Abonour; Courtney E. Bakan; Pascale Andre; Yvonne A. Efebera; Jerome Tiollier; Michael A. Caligiuri; Sherif S. Farag

Natural killer (NK) cells elicit cytotoxicity against multiple myeloma (MM); however, MM cells express HLA class I molecules as ligands to NK cell inhibitory killer immunoglobulin-like receptors (KIRs) as a means of immunoevasion. KIR-ligand mismatch may improve outcomes in allogeneic transplantation for MM. Extrapolating on this concept, we conducted a phase 1 trial of IPH2101, an anti-KIR antibody, in patients with relapsed/refractory MM. IPH2101 was administered intravenously every 28 days in 7 dose-escalated cohorts (0.0003-3 mg/kg) for up to 4 cycles. Pharmacokinetic, pharmacodynamic, and correlative immunologic studies were completed. A total of 32 patients were enrolled. The biologic endpoint of full KIR2D occupancy across the dosing cycle was achieved without dose-limiting toxicity or maximally tolerated dose. One severe adverse event was noted. Pharmacokinetic and pharmacodynamic findings approximated preclinical predictions, and IPH2101 enhanced ex vivo patient-derived NK cell cytotoxicity against MM. No objective responses were seen. No evidence of autoimmunity was observed. These findings suggest that IPH2101 is safe and tolerable at doses that achieve full inhibitory KIR saturation, and this approach warrants further development in MM. This trial was registered at www.clinicaltrials.gov as #NCT00552396.


Cancer Immunology, Immunotherapy | 2013

Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC

Shauna M. Collins; Courtney E. Bakan; Gina D. Swartzel; Craig C. Hofmeister; Yvonne A. Efebera; Hakju Kwon; Gary C. Starling; David Ciarlariello; Shakthi Bhaskar; Edward L. Briercheck; Tiffany Hughes; Jianhua Yu; Audie Rice; Don M. Benson

Abstract Elotuzumab is a monoclonal antibody in development for multiple myeloma (MM) that targets CS1, a cell surface glycoprotein expressed on MM cells. In preclinical models, elotuzumab exerts anti-MM efficacy via natural killer (NK)-cell-mediated antibody-dependent cellular cytotoxicity (ADCC). CS1 is also expressed at lower levels on NK cells where it acts as an activating receptor. We hypothesized that elotuzumab may have additional mechanisms of action via ligation of CS1 on NK cells that complement ADCC activity. Herein, we show that elotuzumab appears to induce activation of NK cells by binding to NK cell CS1 which promotes cytotoxicity against CS1(+) MM cells but not against autologous CS1(+) NK cells. Elotuzumab may also promote CS1–CS1 interactions between NK cells and CS1(+) target cells to enhance cytotoxicity in a manner independent of ADCC. NK cell activation appears dependent on differential expression of the signaling intermediary EAT-2 which is present in NK cells but absent in primary, human MM cells. Taken together, these data suggest elotuzumab may enhance NK cell function directly and confer anti-MM efficacy by means beyond ADCC alone.


Blood | 2011

IPH2101, a novel anti-inhibitory KIR antibody, and lenalidomide combine to enhance the natural killer cell versus multiple myeloma effect.

Don M. Benson; Courtney E. Bakan; Shuhong Zhang; Shauna M. Collins; Jing Liang; Shivani Srivastava; Craig C. Hofmeister; Yvonne A. Efebera; Pascale Andre; Francois Romagne; Mathieu Blery; Cécile Bonnafous; Jianying Zhang; David Clever; Michael A. Caligiuri; Sherif S. Farag

Multiple myeloma (MM) patients who receive killer cell Ig-like receptor (KIR) ligand-mismatched, T cell-depleted, allogeneic transplantation may have a reduced risk of relapse compared with patients who receive KIR ligand-matched grafts, suggesting the importance of this signaling axis in the natural killer (NK) cell-versus-MM effect. Expanding on this concept, IPH2101 (1-7F9), an anti-inhibitory KIR mAb, enhances NK-cell function against autologous MM cells by blocking the engagement of inhibitory KIR with cognate ligands, promoting immune complex formation and NK-cell cytotoxicity specifically against MM cell targets but not normal cells. IPH2101 prevents negative regulatory signals by inhibitory KIR, whereas lenalidomide augments NK-cell function and also appears to up-regulate ligands for activating NK-cell receptors on MM cells. Lenalidomide and a murine anti-inhibitory NK-cell receptor Ab mediate in vivo rejection of a lenalidomide-resistant tumor. These mechanistic, preclinical data support the use of a combination of IPH2101 and lenalidomide in a phase 2 trial for MM.


Clinical Cancer Research | 2015

A phase I trial of the anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma

Don M. Benson; Adam D. Cohen; Sundar Jagannath; Nikhil C. Munshi; Gary Spitzer; Craig C. Hofmeister; Yvonne A. Efebera; Pascale Andre; Robert Albert Zerbib; Michael A. Caligiuri

Purpose: Natural killer (NK) cells may play an important role in the immune response to multiple myeloma; however, multiple myeloma cells express killer immunoglobulin-like receptor (KIR) ligands to prevent NK cell cytotoxicity. Lenalidomide can expand and activate NK cells in parallel with its direct effects against multiple myeloma; however, dexamethasone may impair these favorable immunomodulatory properties. IPH2101, a first-in-class antiinhibitory KIR antibody, has acceptable safety and tolerability in multiple myeloma as a single agent. The present work sought to characterize lenalidomide and IPH2101 as a novel, steroid-sparing, dual immune therapy for multiple myeloma. Experimental Design: A phase I trial enrolled 15 patients in three cohorts. Lenalidomide was administered per os at 10 mg on cohort 1 and 25 mg on cohorts 2 and 3 days 1 to 21 on a 28-day cycle with IPH2101 given intravenously on day 1 of each cycle at 0.2 mg/kg in cohort 1, 1 mg/kg in cohort 2, and 2 mg/kg in cohort 3. No corticosteroids were utilized. The primary endpoint was safety, and secondary endpoints included clinical activity, pharmacokinetics (PK), and pharmacodynamics (PD). Results: The biologic endpoint of full KIR occupancy was achieved across the IPH2101 dosing interval. PD and PK of IPH2101 with lenalidomide were similar to data from a prior single-agent IPH2101 trial. Five serious adverse events (SAE) were reported. Five objective responses occurred. No autoimmunity was seen. Conclusions: These findings suggest that lenalidomide in combination with antiinhibitory KIR therapy warrants further investigation in multiple myeloma as a steroid-sparing, dual immune therapy. This trial was registered at www.clinicaltrials.gov (reference: NCT01217203). Clin Cancer Res; 21(18); 4055–61. ©2015 AACR.


Leukemia & Lymphoma | 2013

Early versus delayed autologous stem cell transplant in patients receiving novel therapies for multiple myeloma

Neil Dunavin; Lai Wei; Patrick Elder; Gary Phillips; Don M. Benson; Craig C. Hofmeister; Sam Penza; Carli N Greenfield; Karen S. Rose; Gisele Rieser; Lisa Merritt; Jill Ketcham; Nyla A. Heerema; John C. Byrd; Steven M. Devine; Yvonne A. Efebera

Abstract Autologous stem cell transplant (ASCT) is an effective treatment for multiple myeloma (MM). However, the timing of ASCT in the era of novel agents (lenalidomide, thalidomide, bortezomib) is unknown. We retrospectively reviewed the outcome of patients with MM who received novel agent-based induction treatment and received first ASCT within 12 months of diagnosis (early ASCT, n = 102) or at a later date (late ASCT, n = 65). Median time to ASCT was 7.9 months vs. 17.7 months in early vs. late ASCT. The 3- and 5-year overall survival (OS) from diagnosis was 90 and 63% vs. 82 and 63% in early and late ASCT, respectively (p = 0.45). Forty-one and 36 patients in the early and late ASCT groups have relapsed or progressed, with median time to relapse of 28 and 23 months (p = 0.055). On multivariable analysis, factors predictive of increased risk for progression were International Scoring System (ISS) stage III (p = 0.007), and less than a very good partial response (< VGPR) post-ASCT (p < 0.001). A factor predictive of worst outcome for OS was being on hemodialysis (p = 0.037). No superiority of one agent was seen. In summary, early or late ASCT is a viable option for patients with MM receiving induction treatment with novel targeted therapies.


The American Journal of Surgical Pathology | 2011

Strong Transthyretin Immunostaining: Potential Pitfall in Cardiac Amyloid Typing

Anjali A. Satoskar; Yvonne A. Efebera; Ayesha Hasan; Sergey V. Brodsky; Gyongyi Nadasdy; Ahmet Dogan; Tibor Nadasdy

Although systemic amyloidosis commonly presents with renal disease, cardiac involvement usually determines the patients prognosis. Cardiac involvement is seen in light chain amyloid and transthyretin amyloidosis. Distinguishing between these two is critical because prognosis and treatment differ. Our study demonstrates the unreliability of transthyretin immunostaining in subtyping cardiac amyloid. Between January 2003 and August 2010, we retrieved 229 native endomyocardial biopsies, of which 24 had amyloid. Immunohistochemistry for &kgr;, &lgr;, transthyretin, and serum amyloid A protein was performed on formalin-fixed, paraffin-embedded sections. Staining was graded as weak (trace to 1+) or strong (2 to 3+). Mass spectrometry (MS)-based proteomic typing of microdissected amyloid material was performed on selected cases. Fifteen patients had monoclonal gammopathy/plasma cell dyscrasia with cardiac amyloid. Eight of them (53%) showed strong transthyretin staining in the cardiac amyloid deposits. MS was performed in 5 of these 8 biopsies, and all 5 biopsies revealed light chain amyloid-type amyloid. Two of these 5 light chain amyloid biopsies did not even have concomitant strong staining for the appropriate light chain. Among the 15 cases with plasma cell dyscrasia, only 7 biopsies showed strong staining for the corresponding monoclonal light chain. Strong, false-positive immunostaining for transthyretin in cardiac amyloid is a potential pitfall, augmented by the frequent lack of staining for immunoglobulin light chains. Therefore, the presence of amyloid in the cardiac biopsy should prompt a search for plasma cell dyscrasia irrespective of transthyretin staining. Confirmation with MS should be sought, particularly if there is any discrepancy between &kgr;/&lgr; staining and serum immunofixation results.


JCI insight | 2017

An early-biomarker algorithm predicts lethal graft-versus-host disease and survival

Matthew J. Hartwell; Umut Ozbek; Ernst Holler; Anne S. Renteria; Hannah Major-Monfried; Pavan Reddy; Mina Aziz; William J. Hogan; Francis Ayuk; Yvonne A. Efebera; Elizabeth O. Hexner; Udomsak Bunworasate; Muna Qayed; Rainer Ordemann; Matthias Wölfl; Stephan Mielke; Attaphol Pawarode; Yi-Bin Chen; Steven M. Devine; Andrew C. Harris; Madan Jagasia; Carrie L. Kitko; Mark R. Litzow; Nicolaus Kröger; Franco Locatelli; George Morales; Ryotaro Nakamura; Ran Reshef; Wolf Rösler; Daniela Weber

BACKGROUND. No laboratory test can predict the risk of nonrelapse mortality (NRM) or severe graft-versus-host disease (GVHD) after hematopoietic cellular transplantation (HCT) prior to the onset of GVHD symptoms. METHODS. Patient blood samples on day 7 after HCT were obtained from a multicenter set of 1,287 patients, and 620 samples were assigned to a training set. We measured the concentrations of 4 GVHD biomarkers (ST2, REG3α, TNFR1, and IL-2Rα) and used them to model 6-month NRM using rigorous cross-validation strategies to identify the best algorithm that defined 2 distinct risk groups. We then applied the final algorithm in an independent test set (n = 309) and validation set (n = 358). RESULTS. A 2-biomarker model using ST2 and REG3α concentrations identified patients with a cumulative incidence of 6-month NRM of 28% in the high-risk group and 7% in the low-risk group (P < 0.001). The algorithm performed equally well in the test set (33% vs. 7%, P < 0.001) and the multicenter validation set (26% vs. 10%, P < 0.001). Sixteen percent, 17%, and 20% of patients were at high risk in the training, test, and validation sets, respectively. GVHD-related mortality was greater in high-risk patients (18% vs. 4%, P < 0.001), as was severe gastrointestinal GVHD (17% vs. 8%, P < 0.001). The same algorithm can be successfully adapted to define 3 distinct risk groups at GVHD onset. CONCLUSION. A biomarker algorithm based on a blood sample taken 7 days after HCT can consistently identify a group of patients at high risk for lethal GVHD and NRM. FUNDING. The National Cancer Institute, American Cancer Society, and the Doris Duke Charitable Foundation.


Biology of Blood and Marrow Transplantation | 2016

International, Multicenter Standardization of Acute Graft-versus-Host Disease Clinical Data Collection: A Report from the Mount Sinai Acute GVHD International Consortium.

Andrew C. Harris; Rachel Young; Steven M. Devine; William J. Hogan; Francis Ayuk; Udomsak Bunworasate; Chantiya Chanswangphuwana; Yvonne A. Efebera; Ernst Holler; Mark R. Litzow; Rainer Ordemann; Muna Qayed; Anne S. Renteria; Ran Reshef; Matthias Wölfl; Yi-Bin Chen; Steven A. Goldstein; Madan Jagasia; Franco Locatelli; Stephan Mielke; David L. Porter; Tal Schechter; Zhanna Shekhovtsova; James L.M. Ferrara; John E. Levine

Acute graft-versus-host disease (GVHD) remains a leading cause of morbidity and nonrelapse mortality after allogeneic hematopoietic cell transplantation. The clinical staging of GVHD varies greatly between transplant centers and is frequently not agreed on by independent reviewers. The lack of standardized approaches to handle common sources of discrepancy in GVHD grading likely contributes to why promising GVHD treatments reported from single centers have failed to show benefit in randomized multicenter clinical trials. We developed guidelines through international expert consensus opinion to standardize the diagnosis and clinical staging of GVHD for use in a large international GVHD research consortium. During the first year of use, the guidance followed discussion of complex clinical phenotypes by experienced transplant physicians and data managers. These guidelines increase the uniformity of GVHD symptom capture, which may improve the reproducibility of GVHD clinical trials after further prospective validation.

Collaboration


Dive into the Yvonne A. Efebera's collaboration.

Researchain Logo
Decentralizing Knowledge