Network


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

Hotspot


Dive into the research topics where Lisa Zimmer is active.

Publication


Featured researches published by Lisa Zimmer.


Science | 2015

Genomic correlates of response to CTLA-4 blockade in metastatic melanoma

Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Sachet A. Shukla; Christian U. Blank; Lisa Zimmer; Antje Sucker; Uwe Hillen; Marnix H. Geukes Foppen; Simone M. Goldinger; Jochen Utikal; Jessica C. Hassel; Benjamin Weide; Katharina C. Kaehler; Carmen Loquai; Peter Mohr; Ralf Gutzmer; Reinhard Dummer; Stacey Gabriel; Catherine J. Wu; Dirk Schadendorf; Levi A. Garraway

Is cancer immunotherapy a private affair? Immune checkpoint blockade, a relatively new cancer treatment, substantially extends the survival of a subset of patients. Previous work has shown that patients whose tumors harbor the largest number of mutations—and thus produce a large number of “neoantigens” recognized as foreign by the immune system—are most likely to benefit. Expanding on these earlier studies, Van Allen et al. studied over 100 patients with melanoma and found a similar correlation (see the Perspective by Gubin and Schreiber). There was no evidence, however, that specific neoantigen sequences were shared by patients who responded. Science, this issue p. 207, see also p. 158 Melanoma patients who respond to immunotherapy do not appear to share common tumor neoantigens. [Also see Perspective by Gubin and Schreiber ] Monoclonal antibodies directed against cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), such as ipilimumab, yield considerable clinical benefit for patients with metastatic melanoma by inhibiting immune checkpoint activity, but clinical predictors of response to these therapies remain incompletely characterized. To investigate the roles of tumor-specific neoantigens and alterations in the tumor microenvironment in the response to ipilimumab, we analyzed whole exomes from pretreatment melanoma tumor biopsies and matching germline tissue samples from 110 patients. For 40 of these patients, we also obtained and analyzed transcriptome data from the pretreatment tumor samples. Overall mutational load, neoantigen load, and expression of cytolytic markers in the immune microenvironment were significantly associated with clinical benefit. However, no recurrent neoantigen peptide sequences predicted responder patient populations. Thus, detailed integrated molecular characterization of large patient cohorts may be needed to identify robust determinants of response and resistance to immune checkpoint inhibitors.


Lancet Oncology | 2012

Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial

Uwe Trefzer; Michael A. Davies; Richard F. Kefford; Paolo Antonio Ascierto; Paul B. Chapman; Igor Puzanov; Axel Hauschild; Caroline Robert; Alain Patrick Algazi; Laurent Mortier; Hussein Tawbi; Tabea Wilhelm; Lisa Zimmer; Julie Switzky; Suzanne Swann; Anne Marie Martin; Mary Guckert; Vicki L. Goodman; Michael R. W. Streit; John M. Kirkwood; Dirk Schadendorf

BACKGROUND Brain metastases are common in patients with metastatic melanoma and median overall survival from their diagnosis is typically 17-22 weeks. We assessed dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain. METHODS We undertook a multicentre, open-label, phase 2 trial in 24 centres in six countries. We enrolled patients with histologically confirmed Val600Glu or Val600Lys BRAF-mutant melanoma and at least one asymptomatic brain metastasis (≥5 mm and ≤40 mm in diameter). Eligible patients were aged 18 years or older, had an Eastern Cooperative Oncology Group performance status of 0 or 1, and had adequate organ function. Patients were split into two cohorts: those in cohort A had not received previous local treatment for brain metastases and those in cohort B had progressive brain metastases after previous local treatments. Patients received 150 mg oral dabrafenib twice a day until disease progression, death, or unacceptable adverse events. The primary endpoint was the proportion of patients with Val600Glu BRAF-mutant melanoma who achieved an overall intracranial response, which was defined as a complete response or partial response assessed with a modified form of Response Evaluation Criteria in Solid Tumors (RECIST 1.1). We included patients who received at least one dose of dabrafenib in efficacy and safety analyses. This study is registered with ClinicalTrials.gov, number NCT01266967. FINDINGS Between Feb 2, 2011, and Aug 5, 2011, we enrolled 172 patients: 89 (52%) in cohort A and 83 (48%) in cohort B. 139 (81%) had Val600Glu BRAF-mutant melanoma. 29 (39·2%, 95% CI 28·0-51·2) of 74 patients with Val600Glu BRAF-mutant melanoma in cohort A achieved an overall intracranial response, as did 20 (30·8%, 19·9-43·4) of 65 in cohort B. One (6·7%, 0·2-31·9) of 15 patients with Val600Lys BRAF-mutant melanoma achieved an overall intracranial response in cohort A, as did four (22·2%, 6·4-47·6) of 18 such patients in cohort B. Treatment-related adverse events of grade 3 or worse occurred in 38 (22%) patients. Eleven (6%) patients developed squamous-cell carcinoma (five [6%] patients in cohort A, of whom one also had keratoacanthoma; six [7%] in cohort B). Four grade 4 treatment-related adverse events occurred in cohort A: one blood amylase increase, one convulsion, one lipase increase, and one neutropenia. Two grade 4 events occurred in cohort B: one agranulocytosis and one intracranial haemorrhage. 51 (30%) patients had a serious adverse event. The three most frequent serious adverse events were pyrexia (ten [6%] patients), intracranial haemorrhage (ten [6%]; one treatment-related), and squamous-cell carcinoma (11 [6%]). INTERPRETATION Dabrafenib has activity and an acceptable safety profile in patients with Val600Glu BRAF-mutant melanoma and brain metastases irrespective of whether they are untreated or have been previously treated but have progressed. FUNDING GlaxoSmithKline.


Journal of Clinical Oncology | 2012

Atypical Melanocytic Proliferations and New Primary Melanomas in Patients With Advanced Melanoma Undergoing Selective BRAF Inhibition

Lisa Zimmer; Uwe Hillen; Elisabeth Livingstone; Mario E. Lacouture; Richard D. Carvajal; Friederike Egberts; Axel Hauschild; Mohammed Kashani-Sabet; Simone M. Goldinger; Reinhard Dummer; Grant A. McArthur; André Scherag; Antje Sucker; Dirk Schadendorf

PURPOSE Selective inhibition of mutant BRAF by using class I RAF inhibitors in patients with metastatic melanoma has resulted in impressive clinical activity. However, there is also evidence that RAF inhibitors might induce carcinogenesis or promote tumor progression via stimulation of MAPK signaling in RAF wild-type cells. We analyzed melanocytic lesions arising under class I RAF inhibitor treatment for dignity, specific genetic mutations, or expression of signal transduction molecules. PATIENTS AND METHODS In all, 22 cutaneous melanocytic lesions that had either developed or considerably changed in morphology in 19 patients undergoing treatment with selective BRAF inhibitors for BRAF-mutant metastatic melanoma at seven international melanoma centers within clinical trials in 2010 and 2011 were analyzed for mutations in BRAF and NRAS genes and immunohistologically assessed for expression of various signal transduction molecules in comparison with 22 common nevi of 21 patients with no history of BRAF inhibitor treatment. RESULTS Twelve newly detected primary melanomas were confirmed in 11 patients within 27 weeks of selective BRAF blockade. In addition, 10 nevi developed of which nine were dysplastic. All melanocytic lesions were BRAF wild type. Explorations revealed that expression of cyclin D1 and pAKT was increased in newly developed primary melanomas compared with nevi (P = .01 and P = .03, respectively). There was no NRAS mutation in common nevi, but BRAF mutations were frequent. CONCLUSION Malignant melanocytic tumors might develop with increased frequency in patients treated with selective BRAF inhibitors supporting a mechanism of BRAF therapy-induced growth and tumorigenesis. Careful surveillance of melanocytic lesions in patients receiving class I RAF inhibitors seems warranted.


European Journal of Cancer | 2016

Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy

Lars Hofmann; Andrea Forschner; Carmen Loquai; Simone M. Goldinger; Lisa Zimmer; Selma Ugurel; Maria I. Schmidgen; Ralf Gutzmer; Jochen Utikal; Daniela Göppner; Jessica C. Hassel; Friedegund Meier; Julia K. Tietze; Ioannis Thomas; Carsten Weishaupt; Martin Leverkus; Renate Wahl; Ursula Dietrich; Claus Garbe; Michael C. Kirchberger; Thomas K. Eigentler; Carola Berking; Anja Gesierich; Angela M. Krackhardt; Dirk Schadendorf; Gerold Schuler; Reinhard Dummer; Lucie Heinzerling

BACKGROUND Anti-programmed cell death receptor-1 (PD-1) antibodies represent an effective treatment option for metastatic melanoma as well as for other cancer entities. They act via blockade of the PD-1 receptor, an inhibitor of the T-cell effector mechanisms that limit immune responses against tumours. As reported for ipilimumab, the anti-PD-1 antibodies pembrolizumab and nivolumab can induce immune-related adverse events (irAEs). These side-effects affect skin, gastrointestinal tract, liver, endocrine system and other organ systems. Since life-threatening and fatal irAEs have been reported, adequate diagnosis and management are essential. METHODS AND FINDINGS In total, 496 patients with metastatic melanoma from 15 skin cancer centers were treated with pembrolizumab or nivolumab; 242 side-effects were described in 138 patients. In 116 of the 138 patients, side-effects affected the skin, gastrointestinal tract, liver, endocrine, and renal system. Rare side-effects included diabetes mellitus, lichen planus, and pancreas insufficiency due to pancreatitis. CONCLUSION Anti-PD1 antibodies can induce a plethora of irAEs. The knowledge of them will allow prompt diagnosis and improve the management resulting in decreased morbidity.


European Journal of Cancer | 2016

Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy

Lisa Zimmer; Simone M. Goldinger; Lars Hofmann; Carmen Loquai; Selma Ugurel; Ioannis Thomas; Maria I. Schmidgen; Ralf Gutzmer; Jochen Utikal; Daniela Göppner; Jessica C. Hassel; Friedegund Meier; Julia K. Tietze; Andrea Forschner; Carsten Weishaupt; Martin Leverkus; Renate Wahl; Ursula Dietrich; Claus Garbe; Michael C. Kirchberger; Thomas K. Eigentler; Carola Berking; Anja Gesierich; Angela M. Krackhardt; Dirk Schadendorf; Gerold Schuler; Reinhard Dummer; Lucie Heinzerling

BACKGROUND Anti-programmed cell death 1 (PD-1) antibodies represent an effective treatment option for metastatic melanoma and other cancer entities. They act via blockade of the PD-1 receptor, an inhibitor of the T-cell effector mechanisms that limit immune responses against tumours. As reported for ipilimumab, the anti-PD-1 antibodies pembrolizumab and nivolumab can induce immune-related adverse events (irAEs). These side-effects can involve skin, gastrointestinal tract, liver, the endocrine system and other organ systems. Since life-threatening and fatal irAEs have been reported, adequate diagnosis and management are essential. METHODS AND FINDINGS In total, 496 patients with metastatic melanoma from 15 skin cancer centres were treated with pembrolizumab or nivolumab. Two hundred forty two side-effects in 138 patients have been analysed. In 77 of the 138 patients side-effects affected the nervous system, respiratory tract, musculoskeletal system, heart, blood and eyes. Not yet reported side-effects such as meningo-(radiculitis), polyradiculitis, cardiac arrhythmia, asystolia, and paresis have been observed. Rare and difficult to manage side-effects such as myasthenia gravis are described in detail. CONCLUSION Anti-PD-1 antibodies can induce a plethora of irAEs. The knowledge of them will allow prompt diagnosis and improve the management resulting in decreased morbidity.


Journal of the National Cancer Institute | 2014

TERT Promoter Mutation Status as an Independent Prognostic Factor in Cutaneous Melanoma

Klaus G. Griewank; Rajmohan Murali; Joan Anton Puig-Butille; Bastian Schilling; Elisabeth Livingstone; Miriam Potrony; Cristina Carrera; Tobias Schimming; Inga Möller; Marion Schwamborn; Antje Sucker; Uwe Hillen; Celia Badenas; Josep Malvehy; Lisa Zimmer; André Scherag; Susana Puig; Dirk Schadendorf

BACKGROUND Recently, TERT promoter mutations were identified at high frequencies in cutaneous melanoma tumor samples and cell lines. The mutations were found to have a UV-signature and to lead to increased TERT gene expression. We analyzed a large cohort of melanoma patients for the presence and distribution of TERT promoter mutations and their association with clinico-pathological characteristics. METHODS 410 melanoma tumor samples were analyzed by Sanger sequencing for the presence of TERT promoter mutations. An analysis of associations between mutation status and various clinical and pathologic variables was performed. RESULTS TERT promoter mutations were identified in 154 (43%) of 362 successfully sequenced melanomas. Mutation frequencies varied between melanoma subtype, being most frequent in melanomas arising in nonacral skin (48%) and melanomas with occult primary (50%), and less frequent in mucosal (23%), and acral (19%) melanomas. Mutations carried a UV signature (C>T or CC>TT). The presence of TERT promoter mutations was associated with factors such as BRAF or NRAS mutation (P < .001), histologic type (P = .002), and Breslow thickness (P < .001). TERT promoter mutation was independently associated with poorer overall survival in patients with nonacral cutaneous melanomas (median survival 80 months vs 291 months for wild-type; hazard ratio corrected for other covariates 2.47; 95% confidence interval [CI] = 1.29 to 4.74; P = .006). CONCLUSIONS UV-induced TERT promoter mutations are one of the most frequent genetic alterations in melanoma, with frequencies varying depending on melanoma subtype. In nonacral cutaneous melanomas, presence of TERT promoter mutations is independently associated with poor prognosis.


European Journal of Cancer | 2015

Acquired BRAF inhibitor resistance: A multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms

Douglas B. Johnson; Alexander M. Menzies; Lisa Zimmer; Zeynep Eroglu; Fei Ye; Shilin Zhao; Helen Rizos; Antje Sucker; Richard A. Scolyer; Ralf Gutzmer; Helen Gogas; Richard F. Kefford; John F. Thompson; Jürgen C. Becker; Carola Berking; Friederike Egberts; Carmen Loquai; Simone M. Goldinger; Gulietta M. Pupo; Willy Hugo; Xiangju Kong; Levi A. Garraway; Jeffrey A. Sosman; Antoni Ribas; Roger S. Lo; Dirk Schadendorf

BACKGROUND Acquired resistance to BRAF inhibitors (BRAFi) is a near-universal phenomenon caused by numerous genetic and non-genetic alterations. In this study, we evaluated the spectrum, onset, pattern of progression, and subsequent clinical outcomes associated with specific mechanisms of resistance. METHODS We compiled clinical and genetic data from 100 patients with 132 tissue samples obtained at progression on BRAFi therapy from 3 large, previously published studies of BRAFi resistance. These samples were subjected to whole-exome sequencing and/or polymerase chain reaction-based genetic testing. RESULTS Among 132 samples, putative resistance mechanisms were identified in 58%, including NRAS or KRAS mutations (20%), BRAF splice variants (16%), BRAF(V600E/K) amplifications (13%), MEK1/2 mutations (7%), and non-mitogen-activated protein kinase pathway alterations (11%). Marked heterogeneity was observed within tumors and patients; 18 of 19 patients (95%) with more than one progression biopsy had distinct/unknown drivers of resistance between samples. NRAS mutations were associated with vemurafenib use (p = 0.045) and intracranial metastases (p = 0.036), and MEK1/2 mutations correlated with hepatic progression (p = 0.011). Progression-free survival and overall survival were similar across resistance mechanisms. The median survival after disease progression was 6.9 months, and responses to subsequent BRAF and MEK inhibition were uncommon (2 of 15; 13%). Post-progression outcomes did not correlate with specific acquired BRAFi-resistance mechanisms. CONCLUSIONS This is the first study to systematically characterise the clinical implications of particular acquired BRAFi-resistance mechanisms in patients with BRAF-mutant melanoma largest study to compile the landscape of resistance. Despite marked heterogeneity of resistance mechanisms within patients, NRAS mutations correlated with vemurafenib use and intracranial disease involvement.


Archives of Dermatology | 2012

Panniculitis with arthralgia in patients with melanoma treated with selective BRAF inhibitors and its management.

Lisa Zimmer; Elisabeth Livingstone; Uwe Hillen; Stephanie D ömkes; Arne Becker; Dirk Schadendorf

BACKGROUND Painful lobular panniculitis appears to be a novel cutaneous adverse effect of selective BRAF inhibitors. OBSERVATION We report the clinical course and management in 2 women with metastatic melanomas harboring the BRAF(V600E) mutation, who developed panniculitis with arthralgia during therapy with selective oral BRAF inhibitors. Panniculitis with arthralgia was the acute presenting adverse effect in both patients. Painful, red, nodular lesions were located on the upper and lower extremities. Biopsy specimens of the nodules showed a mild, predominantly lobular neutrophilic panniculitis. Analgesic and anti-inflammatory treatment improved panniculitis and arthralgia in both cases. It was also necessary to reduce the BRAF inhibitor dose in 1 patient. CONCLUSIONS During therapy with selective BRAF inhibitors, panniculitis with arthralgia represents a new adverse effect that can require dose reduction. In case of this adverse effect, treatment with nonsteroidal anti-inflammatory drugs, such as etoricoxib, should be initiated early to keep patients on treatment and to avoid drug discontinuation and tumor progression.


Nature Immunology | 2011

Failure to detect production of IL-10 by activated human neutrophils.

Martin S. Davey; Nicola Tamassia; Marzia Rossato; Flavia Bazzoni; Federica Calzetti; Kirsten Bruderek; Marina Sironi; Lisa Zimmer; Barbara Bottazzi; Alberto Mantovani; Sven Brandau; Bernhard Moser; Matthias Eberl; Marco A. Cassatella

To the Editor: We read with great interest the paper by De Santo et al.1 published in the November 2010 issue of Nature Immunology, which has received wide attention because of its potential implications for inflammation and immunotherapy. The authors show that the acute-phase protein serum amyloid A1 (SAA-1) induces substantial secretion of the immunosuppressive cytokine interleukin 10 (IL-10) by human neutrophils (up to 100–400 ng/ml) and that invariant natural killer T cells (iNKT cells) are able to diminish this. The authors conclude that harnessing iNKT cells might “be useful therapeutically by decreasing the frequency of immunosuppressive neutrophils and restoring tumor-specific immune responses” in patients with melanoma and other patients with high plasma SAA-1 concentrations1. Intimate crosstalk of neutrophils and monocytes with another population of ‘unconventional’ human T cells, Vg9Vd2 gd T cells, has been demonstrated2. Of relevance for immunotherapy, Vg9Vd2 T cells are much more abundant in human blood than are iNKT cells and can readily be activated by aminobisphosphonate drugs such as zoledronate (Zometa). Small-scale trials have shown promising results for therapy targeting Vg9Vd2 T cells to treat metastatic disease in patients with multiple myeloma, prostate cancer or breast cancer3. Thus, we were keen to ‘translate’ the described interaction of iNKT cells with neutrophils to our own experimental system2,4 and to test whether activated Vg9Vd2 T cells likewise inhibits IL-10-producing neutrophils. Unexpectedly, we found that highly purified, obtained by negative selection human neutrophils (Supplementary Fig. 1) did not secrete IL-10 (Fig. 1a) or express IL-10 mRNA (Fig. 1b) in response to recombinant SAA-1 at the same concentrations used by De Santo et al.1 Notably, and in contrast to De Santo et al., we also did not detect IL-10 in the culture supernatants of neutrophils stimulated with either Salmonella abortus equi lipopolysaccharide (LPS) or ultrapure Escherichia coli LPS (Fig. 1 and Supplementary Fig. 2). The failure to detect IL-10 production by human neutrophils in response to SAA-1 or LPS was reproducible in four independent laboratories at Cardiff (UK), Verona (Italy), Milan (Italy) and Essen (Germany) and was independent of the isolation procedure, the method used to lyse red blood cells, the culture dishes used, cell numbers and medium supplements (Supplementary Methods). The lack of IL-10 in neutrophil culture supernatants was consistent with the absence of both IL-10 mRNA (Fig. 1b) and IL-10 protein (data not shown) in total lysates of LPS-stimulated neutrophils, which confirmed published data5,6. Finally, human neutrophils did not secrete IL-10 even when incubated with other Toll-like receptor agonists (poly(I:C), Pam3CSK4 or R-848), cytokines (IFN-g, TNF, GM-CSF or G-CSF), curdlan, neutrophil-activating protein derived from Helicobacter pylori, the chemoattractant fMLF or insoluble immunocomplexes (Supplementary Fig. 2 and data not shown). In their study, De Santo et al. positively select cells on the basis of CD11b expression and define them as >95% CD11b+CD15+ (ref. 1), compared with the value of over 98–99% achieved in our laboratories2,4,7,8 (Supplementary Methods). Of note, both peripheral neutrophils and monocytes expressed CD11b. Although neutrophils were distinctly CD15+, monocytes had intermediate surface expression of CD15 (Supplementary Fig. 1). Contaminating monocytes may thus give rise to false-positive data in whole-population analyses by enzyme-linked immunosorbent assay (ELISA), quantitative RT-PCR or immunoblot analysis. When we added


PLOS ONE | 2015

Phase II DeCOG-Study of Ipilimumab in Pretreated and Treatment-Naïve Patients with Metastatic Uveal Melanoma

Lisa Zimmer; Julia Vaubel; Peter Mohr; Axel Hauschild; Jochen Utikal; Jan C. Simon; Claus Garbe; Rudolf A. Herbst; Alexander Enk; Eckhart Kämpgen; Elisabeth Livingstone; Leonie Bluhm; Rainer Rompel; Klaus G. Griewank; Michael Fluck; Bastian Schilling; Dirk Schadendorf

Purpose Up to 50% of patients with uveal melanoma (UM) develop metastatic disease with limited treatment options. The immunomodulating agent ipilimumab has shown an overall survival (OS) benefit in patients with cutaneous metastatic melanoma in two phase III trials. As patients with UM were excluded in these studies, the Dermatologic Cooperative Oncology Group (DeCOG) conducted a phase II to assess the efficacy and safety of ipilimumab in patients with metastatic UM. Patients and Methods We undertook a multicenter phase II study in patients with different subtypes of metastatic melanoma. Here we present data on patients with metastatic UM (pretreated and treatment-naïve) who received up to four cycles of ipilimumab administered at a dose of 3 mg/kg in 3 week intervals. Tumor assessments were conducted at baseline, weeks 12, 24, 36 and 48 according to RECIST 1.1 criteria. Adverse events (AEs), including immune-related AEs were graded according to National Cancer Institute Common Toxicity Criteria (CTC) v.4.0. Primary endpoint was the OS rate at 12 months. Results Forty five pretreated (85%) and eight treatment-naïve (15%) patients received at least one dose of ipilimumab. 1-year and 2-year OS rates were 22% and 7%, respectively. Median OS was 6.8 months (95% CI 3.7–8.1), median progression-free survival 2.8 months (95% CI 2.5–2.9). The disease control rate at weeks 12 and 24 was 47% and 21%, respectively. Sixteen patients had stable disease (47%), none experienced partial or complete response. Treatment-related AEs were observed in 35 patients (66%), including 19 grade 3–4 events (36%). One drug-related death due to pancytopenia was observed. Conclusions Ipilimumab has very limited clinical activity in patients with metastatic UM. Toxicity was manageable when treated as per protocol-specific guidelines. Trial Registration ClinicalTrials.gov NCT01355120

Collaboration


Dive into the Lisa Zimmer's collaboration.

Top Co-Authors

Avatar

Dirk Schadendorf

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralf Gutzmer

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antje Sucker

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Jessica C. Hassel

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Selma Ugurel

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Uwe Hillen

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Klaus G. Griewank

University of Duisburg-Essen

View shared research outputs
Researchain Logo
Decentralizing Knowledge