Network


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

Hotspot


Dive into the research topics where Arbel D. Tadmor is active.

Publication


Featured researches published by Arbel D. Tadmor.


Nature | 2015

Mutant MHC class II epitopes drive therapeutic immune responses to cancer

Sebastian Kreiter; Mathias Vormehr; Niels van de Roemer; Mustafa Diken; Martin Löwer; Jan Diekmann; Sebastian Boegel; Barbara Schrörs; Fulvia Vascotto; John C. Castle; Arbel D. Tadmor; Stephen P. Schoenberger; Christoph Huber; Özlem Türeci; Ugur Sahin

Tumour-specific mutations are ideal targets for cancer immunotherapy as they lack expression in healthy tissues and can potentially be recognized as neo-antigens by the mature T-cell repertoire. Their systematic targeting by vaccine approaches, however, has been hampered by the fact that every patient’s tumour possesses a unique set of mutations (‘the mutanome’) that must first be identified. Recently, we proposed a personalized immunotherapy approach to target the full spectrum of a patient’s individual tumour-specific mutations. Here we show in three independent murine tumour models that a considerable fraction of non-synonymous cancer mutations is immunogenic and that, unexpectedly, the majority of the immunogenic mutanome is recognized by CD4+ T cells. Vaccination with such CD4+ immunogenic mutations confers strong antitumour activity. Encouraged by these findings, we established a process by which mutations identified by exome sequencing could be selected as vaccine targets solely through bioinformatic prioritization on the basis of their expression levels and major histocompatibility complex (MHC) class II-binding capacity for rapid production as synthetic poly-neo-epitope messenger RNA vaccines. We show that vaccination with such polytope mRNA vaccines induces potent tumour control and complete rejection of established aggressively growing tumours in mice. Moreover, we demonstrate that CD4+ T cell neo-epitope vaccination reshapes the tumour microenvironment and induces cytotoxic T lymphocyte responses against an independent immunodominant antigen in mice, indicating orchestration of antigen spread. Finally, we demonstrate an abundance of mutations predicted to bind to MHC class II in human cancers as well by employing the same predictive algorithm on corresponding human cancer types. Thus, the tailored immunotherapy approach introduced here may be regarded as a universally applicable blueprint for comprehensive exploitation of the substantial neo-epitope target repertoire of cancers, enabling the effective targeting of every patient’s tumour with vaccines produced ‘just in time’.


Nature | 2017

Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer

Ugur Sahin; Evelyna Derhovanessian; Matthias Miller; Björn-Philipp Kloke; Petra Simon; Martin Löwer; Valesca Bukur; Arbel D. Tadmor; Ulrich Luxemburger; Barbara Schrörs; Tana Omokoko; Mathias Vormehr; Christian Albrecht; Anna Paruzynski; Andreas Kuhn; Janina Buck; Sandra Heesch; Katharina Schreeb; Felicitas Müller; Inga Ortseifer; Isabel Vogler; Eva Godehardt; Sebastian Attig; Richard Rae; Andrea Breitkreuz; Claudia Tolliver; Martin Suchan; Goran Martic; Alexander Hohberger; Patrick Sorn

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of β2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.


BMC Genomics | 2014

Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma

John C. Castle; Martin Loewer; Sebastian Boegel; Jos de Graaf; Christian Bender; Arbel D. Tadmor; Valesca Boisguérin; Thomas Bukur; Patrick Sorn; Claudia Paret; Mustafa Diken; Sebastian Kreiter; Özlem Türeci; Ugur Sahin

BackgroundTumor models are critical for our understanding of cancer and the development of cancer therapeutics. Here, we present an integrated map of the genome, transcriptome and immunome of an epithelial mouse tumor, the CT26 colon carcinoma cell line.ResultsWe found that Kras is homozygously mutated at p.G12D, Apc and Tp53 are not mutated, and Cdkn2a is homozygously deleted. Proliferation and stem-cell markers, including Top2a, Birc5 (Survivin), Cldn6 and Mki67, are highly expressed while differentiation and top-crypt markers Muc2, Ms4a8a (MS4A8B) and Epcam are not. Myc, Trp53 (tp53), Mdm2, Hif1a, and Nras are highly expressed while Egfr and Flt1 are not. MHC class I but not MHC class II is expressed. Several known cancer-testis antigens are expressed, including Atad2, Cep55, and Pbk. The highest expressed gene is a mutated form of the mouse tumor antigen gp70. Of the 1,688 non-synonymous point variations, 154 are both in expressed genes and in peptides predicted to bind MHC and thus potential targets for immunotherapy development. Based on its molecular signature, we predicted that CT26 is refractory to anti-EGFR mAbs and sensitive to MEK and MET inhibitors, as have been previously reported.ConclusionsCT26 cells share molecular features with aggressive, undifferentiated, refractory human colorectal carcinoma cells. As CT26 is one of the most extensively used syngeneic mouse tumor models, our data provide a map for the rationale design of mode-of-action studies for pre-clinical evaluation of targeted- and immunotherapies.


Scientific Reports | 2015

Mutated tumor alleles are expressed according to their DNA frequency

John C. Castle; Martin Loewer; Sebastian Boegel; Arbel D. Tadmor; Valesca Boisguérin; Jos de Graaf; Claudia Paret; Mustafa Diken; Sebastian Kreiter; Özlem Türeci; Ugur Sahin

The transcription of tumor mutations from DNA into RNA has implications for biology, epigenetics and clinical practice. It is not clear if mutations are in general transcribed and, if so, at what proportion to the wild-type allele. Here, we examined the correlation between DNA mutation allele frequency and RNA mutation allele frequency. We sequenced the exome and transcriptome of tumor cell lines with large copy number variations, identified heterozygous single nucleotide mutations and absolute DNA copy number, and determined the corresponding DNA and RNA mutation allele fraction. We found that 99% of the DNA mutations in expressed genes are expressed as RNA. Moreover, we found a high correlation between the DNA and RNA mutation allele frequency. Exceptions are mutations that cause premature termination codons and therefore activate nonsense-mediated decay. Beyond this, we did not find evidence of any wide-scale mechanism, such as allele-specific epigenetic silencing, preferentially promoting mutated or wild-type alleles. In conclusion, our data strongly suggest that genes are equally transcribed from all alleles, mutated and wild-type, and thus transcribed in proportion to their DNA allele frequency.


Cancer Research | 2015

Abstract CT202: IVAC MUTANOME: Individualized vaccines for the treatment of cancer

Bjoern-Philipp Kloke; Cedrik M. Britten; Carmen Loquai; Martin Löwer; Sebastian Attig; Valesca Bukur; Nicole Bidmon; Evelyna Derhovanessian; Jan Diekmann; Mustafa Diken; Angela Filbry; Stephan Grabbe; Sandra Heesch; Christoph Hoeller; David Langer; Uli Luxemburger; Matthias Miller; Felicitas Mueller; Tina Mueller-Brenne; Inga Ortseifer; Burkhard Otte; Anna Paruzynski; Sebastian Petri; Richard Rae; Christine Seck; Kristina Spieß; Arbel D. Tadmor; Jochen Utikal; Klaus Kuehlke; John C. Castle

Cancer arises from the accumulation of genomic alterations and epigenetic changes that constitute a hallmark of cancer. Owing to the molecular heterogeneity in cancer, only a minor fraction of patients profit from approved therapies. Available targeted therapies can only address alterations common to a particular type of cancer and induce transient effects due to the generation of resistant sub-clones. In contrast, the IVAC MUTANOME project aims to immunologically target multiple cancer mutations uniquely expressed in a given patient9s tumor. The IVAC MUTANOME approach should be applicable to the majority of patients irrespective of the tumor entity and offers the potential to exploit the whole tumor mutanome of a given patient using a multi-target approach. The IVAC approach is supported by (i) the availability of technologies that allow fast discovery and validation of individual mutations based on sequencing of whole exome and (ii) an innovative vaccine platform based on RNA-technology supporting fast manufacturing and release of patient-specific vaccines targeting multiple immunogenic mutations within weeks. The phase I study to test the individualized cancer immunotherapeutics for the treatment of malignant melanoma was approved and initiated in 2013 (NCT02035956). With that, the IVAC MUTANOME trial is the first trial in Europe that introduces a fully personalized mutanome vaccine for cancer. The objectives of the clinical trial are to study the feasibility, safety, tolerability and immunogenicity of the IVAC MUTANOME approach for malignant melanoma. Feasibility will be shown by the proven ability to provide the fully personalized IVAC MUANOME vaccine to patients. Recruitment of a patient in the trial repetitively triggers the IVAC MUTANOME process covering (i) the receipt of tumor and blood sample specimens, (ii) the identification, prioritization and confirmation of mutations, (iii) testing of pre-existing immunity against private tumor mutations, (iv) the final selection of mutated sequences, (iv) design, production of a DNA lead structure, (v) GMP manufacturing and release of the patient-specific mRNA, (vi) shipment to the clinical trial site, and (vii) the administration of the IMP to patients. The IVAC MUTANOME recruitment status, manufacturing experiences and treatment status of this first-in-class clinical trial as well as novel data on the immune assessment incl. vaccine-induced mutation-specific T cell responses of the first patients treated will be presented. Citation Format: Bjoern-Philipp Kloke, Cedrik M. Britten, Carmen Loquai, Martin Lower, Sebastian Attig, Valesca Bukur, Nicole Bidmon, Evelyna Derhovanessian, Jan Diekmann, Mustafa Diken, Angela Filbry, Stephan Grabbe, Sandra Heesch, Christoph Hoeller, David Langer, Uli Luxemburger, Matthias Miller, Felicitas Mueller, Tina Mueller-Brenne, Inga Ortseifer, Burkhard Otte, Anna Paruzynski, Sebastian Petri, Richard Rae, Christine Seck, Kristina Spies, Arbel D. Tadmor, Jochen Utikal, Klaus Kuehlke, John Castle, Alexandra Kemmer-Brueck, Isabel Vogler, Andreas N. Kuhn, Sebastian Kreiter, Oezlem Tuereci, Ugur Sahin. IVAC MUTANOME: Individualized vaccines for the treatment of cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT202. doi:10.1158/1538-7445.AM2015-CT202


Nature Biomedical Engineering | 2018

Challenges towards the realization of individualized cancer vaccines

Özlem Türeci; Martin Löwer; Barbara Schrörs; Maren Lang; Arbel D. Tadmor; Ugur Sahin

Bringing truly personalized cancer vaccination with tumour neoantigens to the clinic will require overcoming the challenges of optimized vaccine design, manufacturing and affordability, and identification of the most suitable clinical setting.


Cancer immunology research | 2016

Abstract A110: Mutant MHC class II epitopes drive therapeutic immune responses to cancer

Mathias Vormehr; Sebastian Kreiter; Niels van de Roemer; Mustafa Diken; Martin Löwer; Fulvia Vascotto; Jan Diekmann; Sebastian Boegel; Barbara Schroers; Arbel D. Tadmor; Özlem Türeci; Ugur Sahin

Mutations are regarded as ideal targets for cancer immunotherapy. As neoepitopes with strict lack of expression in any healthy tissue, they are expected to be safe and could bypass the central tolerance mechanisms. Recent advances in nucleic acid sequencing technologies have revolutionized the field of genomics, allowing the readily targeting of mutated neoantigens for personalized cancer vaccination. We demonstrated in three independent murine tumor models that a considerable fraction of non-synonymous cancer mutations is immunogenic and that unexpectedly the immunogenic mutanome is pre-dominantly recognized by CD4+ T cells. RNA vaccination with such MHC class II restricted immunogenic mutations leads to infiltration of CD4+ and CD8+ T cells into the tumor, reduces intratumoral regulatory T cells and ultimately confers strong anti-tumor activity. Encouraged by these findings we set up a process comprising mutation detection by exome sequencing, selection of vaccine targets by solely bioinformatical prioritization of mutated epitopes predicted to be abundantly expressed and presented on MHC class II molecules. Synthetic mRNA vaccines encoding multiple of these prioritized mutated epitopes induce potent tumor control and complete rejection of established aggressively growing tumors in mice. Moreover, we demonstrate that CD4+ T cell neoepitope vaccination primes CTL responses against an independent immunodominant antigen in tumor bearing mice indicating orchestration of antigen spread. Our findings reveal that cancer mutation based MHC class II restricted epitopes are attractive vaccination targets and provide the preclinical proof of concept for an integrated process from tumor sample to a cancer vaccine customized to the unique repertoire of each patient`s tumor. Citation Format: Mathias Vormehr, Sebastian Kreiter, Niels van de Roemer, Mustafa Diken, Martin Lower, Fulvia Vascotto, Jan Diekmann, Sebastian Boegel, Barbara Schroers, Arbel D. Tadmor, Ozlem Tureci, Ugur Sahin. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A110.


Cancer Research | 2016

Abstract CT022: IVAC® MUTANOME - A first-in-human phase I clinical trial targeting individual mutant neoantigens for the treatment of melanoma

Matthias Miller; Carmen Loquai; Björn-Philipp Kloke; Sebastian Attig; Nicole Bidmon; Stefanie Bolte; Valesca Bukur; Evelyna Derhovanessian; Jan Diekmann; Angela Filbry; Sandra Heesch; Christoph Hoeller; Klaus Kuehlcke; David Langer; Martin Loewer; Felicitas Mueller; Inga Ortseifer; Burkhard Otte; Anna Paruzynski; Richard Rae; Barbara Schroers; Christine Seck; Kristina Spiess; Arbel D. Tadmor; Isabel Vogler; Mathias Vormehr; Alexandra Kemmer-Brueck; Andreas Kuhn; Ulrich Luxemburger; Sebastian Kreiter

One of the hallmarks of cancer is the inherent instability of the genome leading to multiple genomic alterations and epigenetic changes that ultimately drive carcinogenesis. These processes lead to a unique molecular profile of every given tumor and to substantial intratumoral heterogeneity of cancer tissues. Recently, a series of independent reports revealed that pre-formed neoantigen specific T-cell responses are of crucial relevance for the clinical efficacy of immune checkpoint inhibitors. However, spontaneous immune recognition of neoantigens seems to be a rare event with only less than 1% of mutations inducing a T-cell response in the tumor-bearing patient. Accordingly, only patients with a high burden of mutations profit from currently approved therapies. To overcome this restriction, the IVAC® MUTANOME-project harnesses the individual patient9s mutation profile by manufacturing highly potent neoantigen-coding RNA vaccines. To this end, the individual mutation repertoire is identified by next-generation-sequencing, potentially immunogenic mutations are selected and incorporated into a poly-epitopic RNA vaccine that is tailored to activate and expand the individual patient9s neoantigen-specific CD4+ and CD8+ T cells. A phase I study to test this novel concept of an active individualized cancer vaccine for the treatment of malignant melanoma was initiated in 2013 (NCT02035956). Notably, BioNTech RNA Pharmaceutical9s IVAC® MUTANOME trial is the first-in-human trial that introduces a fully personalized RNA vaccine for the treatment of malignant melanoma. The objective of this clinical trial is to study the feasibility, safety, tolerability, immunogenicity and the potential clinical activity of the IVAC® MUTANOME approach. The recruitment of a patient into the trial triggers the multi-step IVAC® MUTANOME process covering (i) the receipt and processing of tumor and blood sample specimens, (ii) the identification, prioritization and confirmation of mutations, (iii) testing of pre-existing immunity against identified tumor mutations, (iv) the selection of mutant neoantigen sequences as vaccine targets, (v) design, production of a DNA lead structure, (vi) GMP manufacturing and release of the patient-specific mRNA, (vii) shipment to the clinical trial site and (viii) the administration of the IMP to patients. Detailed information on the trial, the recruitment and treatment status as well as data on the assessment of vaccine induced immune responses will be presented. Citation Format: Matthias Miller, Carmen Loquai, Bjorn-Philipp Kloke, Sebastian Attig, Nicole Bidmon, Stefanie Bolte, Valesca Bukur, Evelyna Derhovanessian, Jan Diekmann, Angela Filbry, Sandra Heesch, Christoph Hoeller, Klaus Kuehlcke, David Langer, Martin Loewer, Felicitas Mueller, Inga Ortseifer, Burkhard Otte, Anna Paruzynski, Richard Rae, Barbara Schroers, Christine Seck, Kristina Spiess, Arbel D. Tadmor, Isabel Vogler, Mathias Vormehr, Alexandra Kemmer-Brueck, Andreas N. Kuhn, Ulrich Luxemburger, Sebastian Kreiter, Jochen Utikal, Stephan Grabbe, Oezlem Tuereci, Ugur Sahin. IVAC® MUTANOME - A first-in-human phase I clinical trial targeting individual mutant neoantigens for the treatment of melanoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr CT022.


Archive | 2016

Predicting t cell epitopes useful for vaccination

Ugur Sahin; Martin Löwer; Arbel D. Tadmor; Sebastian Boegel; Barbara Schrörs; Mathias Vormehr; Sebastian Kreiter


Archive | 2016

PREDICTING IMMUNOGENICITY OF T CELL EPITOPES

Ugur Sahin; Arbel D. Tadmor; John C. Castle; Sebastian Boegel; Martin Löwer

Collaboration


Dive into the Arbel D. Tadmor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge