Network


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

Hotspot


Dive into the research topics where Clare Sun is active.

Publication


Featured researches published by Clare Sun.


Blood | 2015

Partial reconstitution of humoral immunity and fewer infections in patients with chronic lymphocytic leukemia treated with ibrutinib.

Clare Sun; Xin Tian; Yuh Shan Lee; Sreenivasulu Gunti; Andrew Lipsky; Sarah E.M. Herman; Dalia A. Salem; Maryalice Stetler-Stevenson; Constance Yuan; Lela Kardava; Susan Moir; Irina Maric; Janet Valdez; Susan Soto; Gerald Marti; Mohammed Farooqui; Abner Louis Notkins; Adrian Wiestner; Georg Aue

Chronic lymphocytic leukemia (CLL) is characterized by immune dysregulation, often including hypogammaglobulinemia, which contributes to a high rate of infections and morbidity. Ibrutinib, a covalent inhibitor of Bruton tyrosine kinase (BTK), inhibits B-cell receptor signaling and is an effective, US Food and Drug Administration (FDA)-approved treatment of CLL. Inactivating germline mutations in BTK cause a severe B-cell defect and agammaglobulinemia. Therefore, we assessed the impact of ibrutinib on immunoglobulin levels, normal B cells, and infection rate in patients with CLL treated with single-agent ibrutinib on a phase 2 investigator-initiated trial. Consistent with previous reports, immunoglobulin G (IgG) levels remained stable during the first 6 months on treatment, but decreased thereafter. In contrast, there were a transient increase in IgM and a sustained increase in IgA (median increase 45% at 12 months, P < .0001). To distinguish the effects on clonal B cells from normal B cells, we measured serum free light chains (FLCs). In κ-clonal CLL cases, clonal (κ) FLCs were elevated at baseline and normalized by 6 months. Nonclonal (λ) FLCs, which were often depressed at baseline, increased, suggesting the recovery of normal B cells. Consistently, we observed normal B-cell precursors in the bone marrow and an increase in normal B-cell numbers in the peripheral blood. Patients with superior immune reconstitution, as defined by an increase in serum IgA of ≥50% from baseline to 12 months, had a significantly lower rate of infections (P = .03). These data indicate that ibrutinib allows for a clinically meaningful recovery of humoral immune function in patients with CLL. This trial was registered at www.clinicaltrials.gov as #NCT015007330.


Haematologica | 2015

Incidence and risk factors of bleeding-related adverse events in patients with chronic lymphocytic leukemia treated with ibrutinib

Andrew Lipsky; Mohammed Farooqui; Xin Tian; Sabrina Martyr; Ann M. Cullinane; Khanh Nghiem; Clare Sun; Janet Valdez; Carsten U. Niemann; Sarah E. M. Herman; Nakhle S. Saba; Susan Soto; Gerald E. Marti; Gulbu Uzel; Steve M. Holland; Jay N. Lozier; Adrian Wiestner

Ibrutinib is associated with bleeding-related adverse events of grade ≤2 in severity, and infrequently with grade ≥3 events. To investigate the mechanisms of bleeding and identify patients at risk, we prospectively assessed platelet function and coagulation factors in our investigator-initiated trial of single-agent ibrutinib for chronic lymphocytic leukemia. At a median follow-up of 24 months we recorded grade ≤2 bleeding-related adverse events in 55% of 85 patients. No grade ≥3 events occurred. Median time to event was 49 days. The cumulative incidence of an event plateaued by 6 months, suggesting that the risk of bleeding decreases with continued therapy. At baseline, von Willebrand factor and factor VIII levels were often high and normalized on treatment. Platelet function measured via the platelet function analyzer (PFA-100™) was impaired in 22 patients at baseline and in an additional 19 patients on ibrutinib (often transiently). Collagen and adenosine diphosphate induced platelet aggregation was tested using whole blood aggregometry. Compared to normal controls, response to both agonists was decreased in all patients with chronic lymphocytic leukemia, whether on ibrutinib or not. Compared to untreated chronic lymphocytic leukemia patients, response to collagen showed a mild further decrement on ibrutinib, while response to adenosine diphosphate improved. All parameters associated with a significantly increased risk of bleeding-related events were present at baseline, including prolonged epinephrine closure time (HR 2.74, P=0.012), lower levels of von Willebrand factor activity (HR 2.73, P=0.009) and factor VIII (HR 3.73, P=0.0004). In conclusion, both disease and treatment-related factors influence the risk of bleeding. Patients at greater risk for bleeding of grade ≤2 can be identified by clinical laboratory tests and counseled to avoid aspirin, non-steroidal anti-inflammatory drugs and fish oils. ClinicalTrials.gov identifier NCT01500733


Clinical Cancer Research | 2016

Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study

Carsten U. Niemann; Sarah E.M. Herman; Irina Maric; Julio Gomez-Rodriguez; Angélique Biancotto; Betty Y. Chang; Sabrina Martyr; Maryalice Stetler-Stevenson; Constance Yuan; Katherine R. Calvo; Raul C. Braylan; Janet Valdez; Yuh Shan Lee; Deanna H. Wong; Jade Jones; Clare Sun; Gerald E. Marti; Mohammed Farooqui; Adrian Wiestner

Purpose: Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental interactions for proliferation and survival that are at least partially mediated through B-cell receptor (BCR) signaling. Ibrutinib, a Bruton tyrosine kinase inhibitor, disrupts BCR signaling and leads to the egress of tumor cells from the microenvironment. Although the on-target effects on CLL cells are well defined, the impact on the microenvironment is less well studied. We therefore sought to characterize the in vivo effects of ibrutinib on the tumor microenvironment. Experimental Design: Patients received single-agent ibrutinib on an investigator-initiated phase II trial. Serial blood and tissue samples were collected pretreatment and during treatment. Changes in cytokine levels, cellular subsets, and microenvironmental interactions were assessed. Results: Serum levels of key chemokines and inflammatory cytokines decreased significantly in patients on ibrutinib. Furthermore, ibrutinib treatment decreased circulating tumor cells and overall T-cell numbers. Most notably, a reduced frequency of the Th17 subset of CD4+ T cells was observed concurrent with reduced expression of activation markers and PD-1 on T cells. Consistent with direct inhibition of T cells, ibrutinib inhibited Th17 differentiation of murine CD4+ T cells in vitro. Finally, in the bone marrow microenvironment, we found that ibrutinib disaggregated the interactions of macrophages and CLL cells, inhibited secretion of CXCL13, and decreased the chemoattraction of CLL cells. Conclusions: In conjunction with inhibition of BCR signaling, these changes in the tumor microenvironment likely contribute to the antitumor activity of ibrutinib and may impact the efficacy of immunotherapeutic strategies in patients with CLL. Clin Cancer Res; 22(7); 1572–82. ©2015 AACR. See related commentary by Bachireddy and Wu, p. 1547


Blood | 2018

Depth and durability of response to ibrutinib in CLL: 5-year follow-up of a phase II study

Inhye E. Ahn; Mohammed Farooqui; Xin Tian; Janet Valdez; Clare Sun; Susan Soto; Jennifer Lotter; Stephanie Housel; Maryalice Stetler-Stevenson; Constance Yuan; Irina Maric; Katherine R. Calvo; Pia Nierman; Thomas Hughes; Nakhle S. Saba; Gerald E. Marti; Stefania Pittaluga; Sarah E. M. Herman; Carsten U. Niemann; Lone Bredo Pedersen; Christian H Geisler; Richard Childs; Georg Aue; Adrian Wiestner

The safety and efficacy of ibrutinib (420 mg) in chronic lymphocytic leukemia (CLL) were evaluated in a phase 2 study; 51 patients had TP53 aberration (TP53 cohort) and 35 were enrolled because of age 65 years or older (elderly cohort). Both cohorts included patients with treatment-naive (TN) and relapsed/refractory (RR) CLL. With the median follow-up of 4.8 years, 49 (57.0%) of 86 patients remain on study. Treatment was discontinued for progressive disease in 20 (23.3%) patients and for adverse events in 5 (5.8%). Atrial fibrillation occurred in 18 (20.9%) patients for a rate of 6.4 per 100 patient-years. No serious bleeding occurred. The overall response rate at 6 months, the primary study endpoint, was 95.8% for the TP53 cohort (95% confidence interval, 85.7%-99.5%) and 93.9% for the elderly cohort (95% confidence interval, 79.8%-99.3%). Depth of response improved with time: at best response, 14 (29.2%) of 48 patients in the TP53 cohort and 9 (27.3%) of 33 in the elderly cohort achieved a complete response. Median minimal residual disease (MRD) in peripheral blood was 3.8 × 10-2 at 4 years, with MRD-negative (<10-4) remissions in 5 (10.2%) patients. In the TP53 cohort, the estimated 5-year progression-free survival (PFS) was 74.4% in TN-CLL compared with 19.4% in RR-CLL (P = .0002), and overall survival (OS) was 85.3% vs 53.7%, respectively (P = .023). In the elderly cohort, the estimated 5-year PFS and OS in RR-CLL were 64.8% and 71.6%, respectively, and no event occurred in TN-CLL. Long-term administration of ibrutinib was well tolerated and provided durable disease control for most patients. This trial was registered at www.clinicaltrials.gov as #NCT01500733.


Nature Communications | 2017

The evolutionary landscape of chronic lymphocytic leukemia treated with ibrutinib targeted therapy

Dan A. Landau; Clare Sun; Daniel Rosebrock; Sarah E. M. Herman; Joshua Fein; Mariela Sivina; Chingiz Underbayev; Delong Liu; Julia Hoellenriegel; Sarangan Ravichandran; Mohammed Farooqui; Wandi Zhang; Carrie Cibulskis; Asaf Zviran; Donna Neuberg; Dimitri Livitz; Ivana Bozic; Ignaty Leshchiner; Gad Getz; Jan A. Burger; Adrian Wiestner; Catherine J. Wu

Treatment of chronic lymphocytic leukemia (CLL) has shifted from chemo-immunotherapy to targeted agents. To define the evolutionary dynamics induced by targeted therapy in CLL, we perform serial exome and transcriptome sequencing for 61 ibrutinib-treated CLLs. Here, we report clonal shifts (change >0.1 in clonal cancer cell fraction, Q < 0.1) in 31% of patients during the first year of therapy, associated with adverse outcome. We also observe transcriptional downregulation of pathways mediating energy metabolism, cell cycle, and B cell receptor signaling. Known and previously undescribed mutations in BTK and PLCG2, or uncommonly, other candidate alterations are present in seventeen subjects at the time of progression. Thus, the frequently observed clonal shifts during the early treatment period and its potential association with adverse outcome may reflect greater evolutionary capacity, heralding the emergence of drug-resistant clones.In a subset of patients with chronic lymphocytic leukemia (CLL) treated with targeted agents, such as ibrutinib, drug resistant subclones emerge. Here, the authors report on transcriptional changes in CLL patients treated with ibrutinib and identify early clonal shifts associated with evolution of resistant clones.


Haematologica | 2017

Lymphocyte activation gene 3: a novel therapeutic target in chronic lymphocytic leukemia

Mika Shapiro; Yair Herishanu; Ben-Zion Katz; Nili Dezorella; Clare Sun; Sigi Kay; Aaron Polliack; Irit Avivi; Adrian Wiestner; Chava Perry

A novel therapeutic approach in cancer, attempting to stimulate host anti-tumor immunity, involves blocking of immune checkpoints. Lymphocyte activation gene 3 (LAG3) is an immune checkpoint receptor expressed on activated/exhausted T cells. When engaged by the major histocompatibility complex (MHC) class II molecules, LAG3 negatively regulates T-cell function, thereby contributing to tumor escape. Intriguingly, a soluble LAG3 variant activates both immune and malignant MHC class II-presenting cells. In the study herein, we examined the role of LAG3 in the pathogenesis of chronic lymphocytic leukemia, an MHC class II-presenting malignancy, and show that chronic lymphocytic leukemia cells express and secrete LAG3. High levels of surface and soluble LAG3 were associated with the unmutated immunoglobulin variable heavy chain leukemic subtype and a shorter median time from diagnosis to first treatment. Utilizing a mechanism mediated through MHC class II engagement, recombinant soluble LAG3-Ig fusion protein, LAG3-Fc, activated chronic lymphocytic leukemia cells, induced anti-apoptotic pathways and protected the cells from spontaneous apoptosis, effects mediated by SYK, BTK and MAPK signaling. Moreover, LAG3 blocking antibody enhanced in vitro T-cell activation. Our data suggest that soluble LAG3 promotes leukemic cell activation and anti-apoptotic effects through its engagement with MHC class II. Furthermore, MHC class II-presenting chronic lymphocytic leukemia cells may affect LAG3-presenting T cells and impose immune exhaustion on their microenvironment; hence, blocking LAG3-MHC class II interactions is a potential therapeutic target in chronic lymphocytic leukemia.


Blood | 2018

A CD19/CD3 bispecific antibody for effective immunotherapy of chronic lymphocytic leukemia in the ibrutinib era

Hannah R. Robinson; Junpeng Qi; Erika M. Cook; Cydney M. Nichols; Eman L. Dadashian; Chingiz Underbayev; Sarah E. M. Herman; Nakhle S. Saba; Keyvan Keyvanfar; Clare Sun; Inhye E. Ahn; Sivasubramanian Baskar; Christoph Rader; Adrian Wiestner

The Bruton tyrosine kinase inhibitor ibrutinib induces high rates of clinical response in chronic lymphocytic leukemia (CLL). However, there remains a need for adjunct treatments to deepen response and to overcome drug resistance. Blinatumomab, a CD19/CD3 bispecific antibody (bsAb) designed in the BiTE (bispecific T-cell engager) format, is approved by the US Food and Drug Administration for the treatment of relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Because of its short half-life of 2.1 hours, blinatumomab requires continuous intravenous dosing for efficacy. We developed a novel CD19/CD3 bsAb in the single-chain Fv-Fc format (CD19/CD3-scFv-Fc) with a half-life of ∼5 days. In in vitro experiments, both CD19/CD3-scFv-Fc and blinatumomab induced >90% killing of CLL cells from treatment-naïve patients. Antileukemic activity was associated with increased autologous CD8 and CD4 T-cell proliferation, activation, and granzyme B expression. In the NOD/SCID/IL2Rγnull patient-derived xenograft mouse model, once-weekly treatment with CD19/CD3-scFv-Fc eliminated >98% of treatment-naïve CLL cells in blood and spleen. By contrast, blinatumomab failed to induce a response, even when administered daily. We next explored the activity of CD19/CD3-scFv-Fc in the context of ibrutinib treatment and ibrutinib resistance. CD19/CD3-scFv-Fc induced more rapid killing of CLL cells from ibrutinib-treated patients than those from treatment-naïve patients. CD19/CD3-scFv-Fc also demonstrated potent activity against CLL cells from patients with acquired ibrutinib-resistance harboring BTK and/or PLCG2 mutations in vitro and in vivo using patient-derived xenograft models. Taken together, these data support investigation of CD19/CD3 bsAbs and other T cell-recruiting bsAbs as immunotherapies for CLL, especially in combination with ibrutinib or as rescue therapy in ibrutinib-resistant disease.


Leukemia & Lymphoma | 2018

Immunological changes with kinase inhibitor therapy for chronic lymphocytic leukemia

Christopher Pleyer; Adrian Wiestner; Clare Sun

Abstract Ibrutinib and idelalisib are kinase inhibitors that have revolutionized the treatment of chronic lymphocytic leukemia (CLL). Capable of inducing durable remissions, these agents also modulate the immune system. Both ibrutinib and idelalisib abrogate the tumor-supporting microenvironment by disrupting cell–cell interactions, modulating the T-cell compartment, and altering the cytokine milieu. Ibrutinib also partially restores T-cell and myeloid defects associated with CLL. In contrast, immune-related adverse effects, including pneumonitis, colitis, hepatotoxicity, and infections are of particular concern with idelalisib. While opportunistic infections and viral reactivations occur with both ibrutinib and idelalisib, these complications are less common and less severe with ibrutinib, especially when used as monotherapy without additional immunosuppressive agents. This review discusses the impact of ibrutinib and idelalisib on the immune system, including infectious and auto-immune complications as well as their specific effects on the B-cell, T-cell, and myeloid compartment.


Leukemia & Lymphoma | 2018

Polyreactive antibodies in CLL correlate with the level of immunoglobulins not the number of B lymphocytes

Sreenivasulu Gunti; Sarah E. M. Herman; Raju V.S.R.K. Gottumukkala; Ying Xiong; Clare Sun; Gilberto N. Carmona; Adrian Wiestner; Abner Louis Notkins

Sreenivasulu Gunti , Sarah E.M. Herman , Raju V.S.R.K. Gottumukkala†, Ying Xiong, Clare Sun , Gilberto N. Carmona, Adrian Wiestner and Abner L. Notkins Experimental Medicine Section, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA; Hematology Branch, National Institute of Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA


Leukemia & Lymphoma | 2018

Richter transformation to Hodgkin lymphoma on Bruton’s tyrosine kinase inhibitor therapy

Alankrita Taneja; Jade Jones; Stefania Pittaluga; Irina Maric; Mohammed Farooqui; Inhye E. Ahn; Adrian Wiestner; Clare Sun

Alankrita Taneja , Jade Jones , Stefania Pittaluga, Irina Maric, Mohammed Farooqui, Inhye E. Ahn, Adrian Wiestner† and Clare Sun† Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA; Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Laboratory Medicine, Clinical Center National Institutes of Health, Bethesda, MD, USA

Collaboration


Dive into the Clare Sun's collaboration.

Top Co-Authors

Avatar

Adrian Wiestner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mohammed Farooqui

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sarah E. M. Herman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Irina Maric

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Janet Valdez

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xin Tian

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Constance Yuan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Delong Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge