Network


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

Hotspot


Dive into the research topics where Tuna Mutis is active.

Publication


Featured researches published by Tuna Mutis.


Blood | 2016

Daratumumab depletes CD38+ immune-regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma

Jakub Krejcik; Tineke Casneuf; Inger S. Nijhof; Bie Verbist; Jaime Bald; Torben Plesner; Khaja Syed; Kevin Liu; Niels W.C.J. van de Donk; Brendan M. Weiss; Tahamtan Ahmadi; Henk M. Lokhorst; Tuna Mutis; A. Kate Sasser

Daratumumab targets CD38-expressing myeloma cells through a variety of immune-mediated mechanisms (complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and antibody-dependent cellular phagocytosis) and direct apoptosis with crosslinking. These mechanisms may also target nonplasma cells that express CD38, which prompted evaluation of daratumumabs effects on CD38-positive immune subpopulations. Peripheral blood (PB) and bone marrow (BM) from patients with relapsed/refractory myeloma from 2 daratumumab monotherapy studies were analyzed before and during therapy and at relapse. Regulatory B cells and myeloid-derived suppressor cells, previously shown to express CD38, were evaluated for immunosuppressive activity and daratumumab sensitivity in the myeloma setting. A novel subpopulation of regulatory T cells (Tregs) expressing CD38 was identified. These Tregs were more immunosuppressive in vitro than CD38-negative Tregs and were reduced in daratumumab-treated patients. In parallel, daratumumab induced robust increases in helper and cytotoxic T-cell absolute counts. In PB and BM, daratumumab induced significant increases in CD8(+):CD4(+) and CD8(+):Treg ratios, and increased memory T cells while decreasing naïve T cells. The majority of patients demonstrated these broad T-cell changes, although patients with a partial response or better showed greater maximum effector and helper T-cell increases, elevated antiviral and alloreactive functional responses, and significantly greater increases in T-cell clonality as measured by T-cell receptor (TCR) sequencing. Increased TCR clonality positively correlated with increased CD8(+) PB T-cell counts. Depletion of CD38(+) immunosuppressive cells, which is associated with an increase in T-helper cells, cytotoxic T cells, T-cell functional response, and TCR clonality, represents possible additional mechanisms of action for daratumumab and deserves further exploration.


Immunological Reviews | 2016

Monoclonal antibodies targeting CD38 in hematological malignancies and beyond

Niels W.C.J. van de Donk; Maarten L. Janmaat; Tuna Mutis; Jeroen J. Lammerts van Bueren; Tahamtan Ahmadi; A. Kate Sasser; Henk M. Lokhorst; Paul Parren

CD38 is a multifunctional cell surface protein that has receptor as well as enzyme functions. The protein is generally expressed at low levels on various hematological and solid tissues, while plasma cells express particularly high levels of CD38. The protein is also expressed in a subset of hematological tumors, and shows especially broad and high expression levels in plasma cell tumors such as multiple myeloma (MM). Together, this triggered the development of various therapeutic CD38 antibodies, including daratumumab, isatuximab, and MOR202. Daratumumab binds a unique CD38 epitope and showed strong anti‐tumor activity in preclinical models. The antibody engages diverse mechanisms of action, including complement‐dependent cytotoxicity, antibody‐dependent cellular cytotoxicity, antibody‐dependent cellular phagocytosis, programmed cell death, modulation of enzymatic activity, and immunomodulatory activity. CD38‐targeting antibodies have a favorable toxicity profile in patients, and early clinical data show a marked activity in MM, while studies in other hematological malignancies are ongoing. Daratumumab has single agent activity and a limited toxicity profile, allowing favorable combination therapies with existing as well as emerging therapies, which are currently evaluated in the clinic. Finally, CD38 antibodies may have a role in the treatment of diseases beyond hematological malignancies, including solid tumors and antibody‐mediated autoimmune diseases.


Haematologica | 2016

Pre-clinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma

Esther Drent; Richard W.J. Groen; Willy Noort; Maria Themeli; Jeroen J. Lammerts van Bueren; Paul Parren; Jürgen Kuball; Zsolt Sebestyen; Huipin Yuan; Joost de de Bruijn; Niels W.C.J. van de Donk; Anton Martens; Henk M. Lokhorst; Tuna Mutis

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we transduced T cells from healthy donors and multiple myeloma patients. We then evaluated the preclinical efficacy and safety of the transduced T cells. Irrespective of the donor and antibody sequence, CD38-chimeric antigen receptor-transduced T cells proliferated, produced inflammatory cytokines and effectively lysed malignant cell lines and primary malignant cells from patients with acute myeloid leukemia and multi-drug resistant multiple myeloma in a cell-dose, and CD38-dependent manner, despite becoming CD38-negative during culture. CD38-chimeric antigen receptor-transduced T cells also displayed significant anti-tumor effects in a xenotransplant model, in which multiple myeloma tumors were grown in a human bone marrow-like microenvironment. CD38-chimeric antigen receptor-transduced T cells also appeared to lyse the CD38+ fractions of CD34+ hematopoietic progenitor cells, monocytes, natural killer cells, and to a lesser extent T and B cells but did not inhibit the outgrowth of progenitor cells into various myeloid lineages and, furthermore, were effectively controllable with a caspase-9-based suicide gene. These results signify the potential importance of CD38-chimeric antigen receptor-transduced T cells as therapeutic tools for CD38+ malignancies and warrant further efforts to diminish the undesired effects of this immunotherapy using appropriate strategies.


Clinical Cancer Research | 2017

Monocytes and granulocytes reduce CD38 expression levels on myeloma cells in patients treated with daratumumab

Jakub Krejcik; Kris A. Frerichs; Inger S. Nijhof; Berris van Kessel; Jeroen F. van Velzen; Andries C. Bloem; Marloes E.C. Broekmans; Sonja Zweegman; Johan van Meerloo; René J. P. Musters; Pino J. Poddighe; Richard W.J. Groen; Christopher Chiu; Torben Plesner; Henk M. Lokhorst; A. Kate Sasser; Tuna Mutis; Niels W.C.J. van de Donk

Purpose: Daratumumab treatment results in a marked reduction of CD38 expression on multiple myeloma cells. The aim of this study was to investigate the clinical implications and the underlying mechanisms of daratumumab-mediated CD38 reduction. Experimental Design: We evaluated the effect of daratumumab alone or in combination with lenalidomide-dexamethasone, on CD38 levels of multiple myeloma cells and nontumor immune cells in the GEN501 study (daratumumab monotherapy) and the GEN503 study (daratumumab combined with lenalidomide-dexamethasone). In vitro assays were also performed. Results: In both trials, daratumumab reduced CD38 expression on multiple myeloma cells within hours after starting the first infusion, regardless of depth and duration of the response. In addition, CD38 expression on nontumor immune cells, including natural killer cells, T cells, B cells, and monocytes, was also reduced irrespective of alterations in their absolute numbers during therapy. In-depth analyses revealed that CD38 levels of multiple myeloma cells were only reduced in the presence of complement or effector cells, suggesting that the rapid elimination of CD38high multiple myeloma cells can contribute to CD38 reduction. In addition, we discovered that daratumumab–CD38 complexes and accompanying cell membrane were actively transferred from multiple myeloma cells to monocytes and granulocytes. This process of trogocytosis was also associated with reduced surface levels of some other membrane proteins, including CD49d, CD56, and CD138. Conclusions: Daratumumab rapidly reduced CD38 expression levels, at least in part, through trogocytosis. Importantly, all these effects also occurred in patients with deep and durable responses, thus excluding CD38 reduction alone as a mechanism of daratumumab resistance. The trials were registered at www.clinicaltrials.gov as NCT00574288 (GEN501) and NCT1615029 (GEN503). Clin Cancer Res; 23(24); 7498–511. ©2017 AACR.


Molecular Therapy | 2017

A Rational Strategy for Reducing On-Target Off-Tumor Effects of CD38-Chimeric Antigen Receptors by Affinity Optimization

Esther Drent; Maria Themeli; Renée Poels; Regina de Jong-Korlaar; Huipin Yuan; Joost D. de Bruijn; Anton Martens; Sonja Zweegman; Niels W.C.J. van de Donk; Richard W.J. Groen; Henk M. Lokhorst; Tuna Mutis

Chimeric antigen receptors (CARs) can effectively redirect cytotoxic Txa0cells toward highly expressed surface antigens on tumor cells. The low expression of several tumor-associated antigens (TAAs) on normal tissues, however, hinders their safe targeting by CAR Txa0cells due to on-target/off-tumor effects. Using the multiple myeloma (MM)-associated CD38 antigen as a model system, here, we present a rational approach for effective and tumor-selective targeting of such TAAs. Using light-chain exchange technology, we combined the heavy chains of two high-affinity CD38 antibodies with 176 germline light chains and generated ∼124 new antibodies with 10- to >1,000-fold lower affinities to CD38. After categorizing them into three distinct affinity classes, we incorporated the single-chain variable fragments of eight antibodies from each class into new CARs. Txa0cells carrying these CD38-CARs were extensively evaluated for their on-tumor/off-tumor cytotoxicity as well as CD38-dependent proliferation and cytokine production. We identified CD38-CAR Txa0cells of ∼1,000- fold reduced affinity, which optimally proliferated, produced Th1-like cytokines, and effectively lysed CD382+ MM cells, but spared CD38+ healthy hematopoietic cells inxa0vitro and inxa0vivo. Thus, this systematicxa0approach is highly suitable for the generation of optimal CARs for effective and selective targeting of TAAs.


Blood | 2016

Phase 1/2 study of lenalidomide combined with low-dose cyclophosphamide and prednisone in lenalidomide-refractory multiple myeloma.

Inger S. Nijhof; Laurens E. Franssen; Mark-David Levin; Gerard M. J. Bos; Annemiek Broijl; Saskia K. Klein; Harry R. Koene; Andries C. Bloem; Aart Beeker; Laura M. Faber; Ellen van der Spek; Paula F. Ypma; Reinier Raymakers; Dick Johan van Spronsen; Peter E. Westerweel; Rimke Oostvogels; Jeroen F. van Velzen; Berris van Kessel; Tuna Mutis; Pieter Sonneveld; Sonja Zweegman; Henk M. Lokhorst; Niels W.C.J. van de Donk

The prognosis of multiple myeloma (MM) patients who become refractory to lenalidomide and bortezomib is very poor, indicating the need for new therapeutic strategies for these patients. Next to the development of new drugs, the strategy of combining agents with synergistic activity may also result in clinical benefit for patients with advanced myeloma. We have previously shown in a retrospective analysis that lenalidomide combined with continuous low-dose cyclophosphamide and prednisone (REP) had remarkable activity in heavily pretreated, lenalidomide-refractory MM patients. To evaluate this combination prospectively, we initiated a phase 1/2 study to determine the optimal dose and to assess its efficacy and safety in lenalidomide-refractory MM patients. The maximum tolerated dose (MTD) was defined as 25 mg lenalidomide (days 1-21/28 days), combined with continuous cyclophosphamide (50 mg/d) and prednisone (20 mg/d). At the MTD (n = 67 patients), the overall response rate was 67%, and at least minimal response was achieved in 83% of the patients. Median progression-free survival and overall survival were 12.1 and 29.0 months, respectively. Similar results were achieved in the subset of patients with lenalidomide- and bortezomib-refractory disease as well as in patients with high-risk cytogenetic abnormalities, defined as t(4;14), t(14;16), del(17p), and/or ampl(1q) as assessed by fluorescence in situ hybridization. Neutropenia (22%) and thrombocytopenia (22%) were the most common grade 3-4 hematologic adverse events. Infections (21%) were the most common grade 3-5 nonhematologic adverse events. In conclusion, the addition of continuous low-dose oral cyclophosphamide to lenalidomide and prednisone offers a new therapeutic perspective for multidrug refractory MM patients. This trial was registered at www.clinicaltrials.gov as #NCT01352338.


European Journal of Haematology | 2016

Outcome of allogeneic transplantation in newly diagnosed and relapsed/refractory multiple myeloma: long-term follow-up in a single institution

Laurens E. Franssen; Reinier Raymakers; Arjan Buijs; Marian F. Schmitz; Suzanne van Dorp; Tuna Mutis; Henk M. Lokhorst; Niels W.C.J. van de Donk

Allogeneic stem cell transplantation (allo‐SCT) has the potential to induce long‐term remission in multiple myeloma (MM), but the role of allo‐SCT in MM is controversial due to the high rate of treatment‐related mortality (TRM). However, although proteasome inhibitors and immunomodulatory drugs have improved the outcome of patients with MM, high‐risk patients still have a very poor prognosis. This indicates the need for new treatment strategies and identification of patients who might benefit from allo‐SCT. We therefore analyzed the outcome of one hundred and forty‐seven patients with MM who received an allo‐SCT at our institution (58 in first line, 89 in relapsed/refractory setting) after a median follow‐up of 88.8 months. For the first‐line setting, median progression‐free survival (PFS) and overall survival (OS) were remarkably good, with a CR rate of 48.3%, median PFS of 30.2 months, and 10‐yr OS of 51%. We found no difference in outcome for patients with high‐risk metaphase cytogenetics or FISH del(13q14), but efficacy in current standard high‐risk patients could not be determined. The outcome in the relapsed/refractory setting was poor, especially in the subgroup of patients relapsing within 18 months after auto‐SCT. Therefore, if applied at all in these patients, improvement of allo‐SCT is needed, focusing on reduction of TRM and more effective immunotherapy.


Journal for ImmunoTherapy of Cancer | 2015

Preclinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma

Esther Drent; Richard W.J. Groen; Willy Noort; Jeroen J. Lammerts van Bueren; Paul Parren; Jürgen Kuball; Zsolt Sebestyen; Niels W.C.J. van de Donk; Anton Martens; Henk Lokhorst; Tuna Mutis

Background Adoptive transfer of T cells transduced with tumorreactive Chimeric Antigen Receptors (CARs) is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high and homogenous expression on Multiple Myeloma (MM) cells, appears a suitable target for antibody therapy. Prompted by this, we evaluated the feasibility of targeting MM with CD38-CAR-transduced (CD38-CAR) T cells.


Blood | 2015

HY antibodies as biomarkers for chronic GVHD

Tuna Mutis

In this issue of Blood , Nakasone et al report on an early biomarker for chronic graft-versus-host disease (cGVHD). 1 In a prospective study, they demonstrated that the detection of multiple HY antibodies (HY-Abs) 3 months after female-to-male allogeneic hematopoietic cell transplant (allo-HCT) can predict cGVHD development, severity, and nonrelapse mortality (NRM). 1


Haematologica | 2018

Cereblon loss and up-regulation of c-Myc are associated with lenalidomide resistance in multiple myeloma patients

Laurens E. Franssen; Inger S. Nijhof; Suzana Couto; Mark-David Levin; Gerard M. J. Bos; Annemiek Broijl; Saskia K. Klein; Yan Ren; Maria Wang; Harry R. Koene; Andries C. Bloem; Aart Beeker; Laura M. Faber; Ellen van der Spek; Reinier Raymakers; Roos J. Leguit; Pieter Sonneveld; Sonja Zweegman; Henk M. Lokhorst; Tuna Mutis; Anjan Thakurta; Xiaozhong Qian; Niels W.C.J. van de Donk

Multiple myeloma (MM) patients who become refractory to anti-MM drugs have a very poor prognosis. Therefore, it is important to gain insight into the mechanisms of resistance to these drugs. Immunomodulatory drugs (IMiDs) have immune-stimulatory and anti-angiogenic properties as well as direct anti-

Collaboration


Dive into the Tuna Mutis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Henk M. Lokhorst

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard W.J. Groen

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sonja Zweegman

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Inger S. Nijhof

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Maria Themeli

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Esther Drent

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anton Martens

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Torben Plesner

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge