Network


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

Hotspot


Dive into the research topics where Simone M. Goldinger is active.

Publication


Featured researches published by Simone M. Goldinger.


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.


PLOS ONE | 2013

The price of tumor control: an analysis of rare side effects of anti-CTLA-4 therapy in metastatic melanoma from the ipilimumab network

Caroline J. Voskens; Simone M. Goldinger; Carmen Loquai; Caroline Robert; Katharina C. Kaehler; Carola Berking; Tanja Bergmann; Clemens L. Bockmeyer; Thomas K. Eigentler; Michael Fluck; Claus Garbe; Ralf Gutzmer; Stephan Grabbe; Axel Hauschild; Rüdiger Hein; Gheorghe Hundorfean; Armin Justich; Ullrich Keller; Christina Klein; C. Mateus; Peter Mohr; Sylvie Paetzold; Imke Satzger; Dirk Schadendorf; Marc Schlaeppi; Gerold Schuler; Beatrice Schuler-Thurner; Uwe Trefzer; Jens Ulrich; Julia Vaubel

Background Ipilimumab, a cytotoxic T-lymphocyte antigen-4 (CTLA-4) blocking antibody, has been approved for the treatment of metastatic melanoma and induces adverse events (AE) in up to 64% of patients. Treatment algorithms for the management of common ipilimumab-induced AEs have lead to a reduction of morbidity, e.g. due to bowel perforations. However, the spectrum of less common AEs is expanding as ipilimumab is increasingly applied. Stringent recognition and management of AEs will reduce drug-induced morbidity and costs, and thus, positively impact the cost-benefit ratio of the drug. To facilitate timely identification and adequate management data on rare AEs were analyzed at 19 skin cancer centers. Methods and Findings Patient files (n = 752) were screened for rare ipilimumab-associated AEs. A total of 120 AEs, some of which were life-threatening or even fatal, were reported and summarized by organ system describing the most instructive cases in detail. Previously unreported AEs like drug rash with eosinophilia and systemic symptoms (DRESS), granulomatous inflammation of the central nervous system, and aseptic meningitis, were documented. Obstacles included patientś delay in reporting symptoms and the differentiation of steroid-induced from ipilimumab-induced AEs under steroid treatment. Importantly, response rate was high in this patient population with tumor regression in 30.9% and a tumor control rate of 61.8% in stage IV melanoma patients despite the fact that some patients received only two of four recommended ipilimumab infusions. This suggests that ipilimumab-induced antitumor responses can have an early onset and that severe autoimmune reactions may reflect overtreatment. Conclusion The wide spectrum of ipilimumab-induced AEs demands doctor and patient awareness to reduce morbidity and treatment costs and true ipilimumab success is dictated by both objective tumor responses and controlling severe side effects.


European Journal of Cancer | 2014

Vemurafenib in patients with BRAFV600 mutation-positive melanoma with symptomatic brain metastases: Final results of an open-label pilot study

Reinhard Dummer; Simone M. Goldinger; Christian P. Turtschi; Nina Eggmann; Olivier Michielin; Lada Mitchell; Luisa Veronese; Paul René Hilfiker; Lea Felderer; Jeannine D. Rinderknecht

BACKGROUND & AIM Brain metastases are frequent in patients with metastatic melanoma, indicating poor prognosis. We investigated the BRAF kinase inhibitor vemurafenib in patients with advanced melanoma with symptomatic brain metastases. METHODS This open-label trial assessed vemurafenib (960mg twice a day) in patients with BRAF(V600) mutation-positive metastatic melanoma with non-resectable, previously treated brain metastases. The primary end-point was safety. Secondary end-points included best overall response rate, and progression-free and overall survival. RESULTS Twenty-four patients received vemurafenib for a median treatment duration of 3.8 (0.1-11.3) months. The majority of discontinuations were due to disease progression (n=22). Twenty-three of 24 patients reported at least one adverse event (AE). Grade 3 AEs were reported in four (17%; 95% confidence interval [CI], 4.7-37.4%) patients and included cutaneous squamous cell carcinoma in four patients. Median progression-free survival was 3.9 (95% CI, 3.0-5.5) months, and median survival was 5.3 (95% CI, 3.9-6.6) months. An overall partial response (PR) at both intracranial and extracranial sites was achieved in 10 of 24 (42%; 95% CI, 22.1-63.4) evaluable patients, with stable disease in nine (38%; 95% CI, 18.8-59.4) patients. Of 19 patients with measurable intracranial disease, seven (37%) achieved >30% intracranial tumour regression, and three (16%; 95% CI, 3.4-39.6%) achieved a confirmed PR. Other signs of improvement included reduced need for corticosteroids and enhanced performance status. CONCLUSIONS Vemurafenib can be safely used in patients with advanced symptomatic melanoma that has metastasised to the brain and can result in meaningful tumour regression.


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.


The New England Journal of Medicine | 2012

Ultraviolet A and Photosensitivity during Vemurafenib Therapy

Reinhard Dummer; Jeannine D. Rinderknecht; Simone M. Goldinger

The BRAF inhibitor, vemurafenib, is associated with photosensitivity. This small study identifies ultraviolet A (UVA) as the active agent. UVA blockers may be effective in preventing photosensitivity among patients taking vemurafenib.


European Journal of Immunology | 2012

Nano-particle vaccination combined with TLR-7 and -9 ligands triggers memory and effector CD8⁺ T-cell responses in melanoma patients

Simone M. Goldinger; Reinhard Dummer; Petra Baumgaertner; Daniela Mihic-Probst; Katrin Schwarz; Anya Hammann-Haenni; Joerg Willers; Christine Geldhof; John O. Prior; Thomas M. Kündig; Olivier Michielin; Martin F. Bachmann; Daniel E. Speiser

Optimal vaccine strategies must be identified for improving T‐cell vaccination against infectious and malignant diseases. MelQbG10 is a virus‐like nano‐particle loaded with A‐type CpG‐oligonucleotides (CpG‐ODN) and coupled to peptide16–35 derived from Melan‐A/MART‐1. In this phase IIa clinical study, four groups of stage III‐IV melanoma patients were vaccinated with MelQbG10, given (i) with IFA (Montanide) s.c.; (ii) with IFA s.c. and topical Imiquimod; (iii) i.d. with topical Imiquimod; or (iv) as intralymph node injection. In total, 16/21 (76%) patients generated ex vivo detectable Melan‐A/MART‐1‐specific T‐cell responses. T‐cell frequencies were significantly higher when IFA was used as adjuvant, resulting in detectable T‐cell responses in all (11/11) patients, with predominant generation of effector‐memory‐phenotype cells. In turn, Imiquimod induced higher proportions of central‐memory‐phenotype cells and increased percentages of CD127+ (IL‐7R) T cells. Direct injection of MelQbG10 into lymph nodes resulted in lower T‐cell frequencies, associated with lower proportions of memory and effector‐phenotype T cells. Swelling of vaccine site draining lymph nodes, and increased glucose uptake at PET/CT was observed in 13/15 (87%) of evaluable patients, reflecting vaccine triggered immune reactions in lymph nodes. We conclude that the simultaneous use of both Imiquimod and CpG‐ODN induced combined memory and effector CD8+ T‐cell responses.


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.


PLOS ONE | 2013

RASopathic Skin Eruptions during Vemurafenib Therapy

Jeannine D. Rinderknecht; Simone M. Goldinger; Sima Rozati; Jivko Kamarashev; Katrin Kerl; Lars E. French; Reinhard Dummer; Benedetta Belloni

Purpose Vemurafenib is a potent inhibitor of V600 mutant BRAF with significant impact on progression-free and overall survival in advanced melanoma. Cutaneous side effects are frequent. This single-center observational study investigates clinical and histological features of these class-specific cutaneous adverse reactions. Patients and Methods Patients were all treated with Vemurafenib 960 mg b.i.d. within local ethic committees approved clinical trials. All skin reactions were collected and documented prospectively. Cutaneous reactions were classified by reaction pattern as phototoxic and inflammatory, hair and nail changes, keratinocytic proliferations and melanocytic disorders. Results Vemurafenib was well tolerated, only in two patients the dose had to be reduced to 720 mg due to arthralgia. 26/28 patients (93%) experienced cutaneous side effects. Observed side effects included UVA dependent photosensitivity (n = 16), maculopapular exanthema (n = 14), pruritus (n = 8), folliculitis (n = 5), burning feet (n = 3), hair thinning (mild alopecia) (n = 8), curly hair (n = 2) and nail changes (n = 2). Keratosis pilaris and acanthopapilloma were common skin reactions (n = 12/n = 13), as well as plantar hyperkeratosis (n = 4), keratoacanthoma (n = 5) and invasive squamous cell carcinoma (n = 4). One patient developed a second primary melanoma after more than 4 months of therapy (BRAF and RAS wild type). Conclusion Vemurafenib has a broad and peculiar cutaneous side effect profile involving epidermis and adnexa overlapping with the cutaneous manifestations of genetic diseases characterized by activating germ line mutations of RAS (RASopathy). They must be distinguished from allergic drug reaction. Regular skin examination and management by experienced dermatologists as well as continuous prophylactic photo protection including an UVA optimized sun screen is mandatory.

Collaboration


Dive into the Simone M. Goldinger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Schadendorf

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Olivier Michielin

University Hospital of Lausanne

View shared research outputs
Top Co-Authors

Avatar

Lisa Zimmer

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Ralf Gutzmer

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge