Network


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

Hotspot


Dive into the research topics where Pia Kvistborg is active.

Publication


Featured researches published by Pia Kvistborg.


Science | 2015

Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer

Naiyer A. Rizvi; Matthew D. Hellmann; Alexandra Snyder; Pia Kvistborg; Vladimir Makarov; Jonathan J. Havel; William R. Lee; Jianda Yuan; Phillip Wong; Teresa S. Ho; Martin L. Miller; Natasha Rekhtman; Andre L. Moreira; Fawzia Ibrahim; Cameron Bruggeman; Billel Gasmi; Roberta Zappasodi; Yuka Maeda; Chris Sander; Edward B. Garon; Taha Merghoub; Jedd D. Wolchok; Ton N. M. Schumacher; Timothy A. Chan

Immune checkpoint inhibitors, which unleash a patient’s own T cells to kill tumors, are revolutionizing cancer treatment. To unravel the genomic determinants of response to this therapy, we used whole-exome sequencing of non–small cell lung cancers treated with pembrolizumab, an antibody targeting programmed cell death-1 (PD-1). In two independent cohorts, higher nonsynonymous mutation burden in tumors was associated with improved objective response, durable clinical benefit, and progression-free survival. Efficacy also correlated with the molecular smoking signature, higher neoantigen burden, and DNA repair pathway mutations; each factor was also associated with mutation burden. In one responder, neoantigen-specific CD8+ T cell responses paralleled tumor regression, suggesting that anti–PD-1 therapy enhances neoantigen-specific T cell reactivity. Our results suggest that the genomic landscape of lung cancers shapes response to anti–PD-1 therapy. An anticancer drug is more effective against tumors that carry more mutations. More mutations predict better efficacy Despite the remarkable success of cancer immunotherapies, many patients do not respond to treatment. Rizvi et al. studied the tumors of patients with non–small-cell lung cancer undergoing immunotherapy. In two independent cohorts, treatment efficacy was associated with a higher number of mutations in the tumors. In one patient, a tumor-specific T cell response paralleled tumor regression. Science, this issue p. 124


Journal of Clinical Oncology | 2013

Tumor Exome Analysis Reveals Neoantigen-Specific T-Cell Reactivity in an Ipilimumab-Responsive Melanoma

Nienke van Rooij; Marit M. van Buuren; Daisy Philips; Arno Velds; Mireille Toebes; Bianca Heemskerk; Laura van Dijk; Sam Behjati; Henk Hilkmann; Dris El Atmioui; Marja Nieuwland; Michael R. Stratton; Ron M. Kerkhoven; Can Keşmir; John B. A. G. Haanen; Pia Kvistborg; Ton N. M. Schumacher

The evidence for T-cell–mediated regression of human cancers such as non–small-cell lung carcinoma, renal cell carcinoma, and—in particular—melanoma after immunotherapy is strong. Anti-CTLA4 (ipilimumab) treatment has been approved for treatment of meta-static melanoma,1 and antibody-mediated blockade of PD-1, a second inhibitory receptor on T cells, has shown highly encouraging results in early clinical trials.2,3 Although the clinical activity of these treatments is apparent, it is still unknown which T-cell reactivities are involved in immunotherapy-induced cancer regression.4 T-cell reactivity against nonmutated tumor-associated self-antigens has been analyzed in patients treated with ipilimumab or with autologous tumor-infiltrating T cells, but the magnitude of the T-cell responses observed has been relatively modest.5,6 In part on the basis of such data, recognition of patient-specific mutant epitopes (hereafter referred to as neoantigens) has been suggested to be a potentially important component.7 A potential involvement of mutated epitopes in T-cell control would also fit well with the observation that the mutation load in sun-exposed melanomas is particularly high.8-10 Intriguingly, on the basis of animal model data, it has recently been suggested that (therapy-induced) analysis of T-cell reactivity against patient-specific neoantigens may be feasible through exploitation of cancer genome data.11,12 However, human data have thus far been lacking. Here we report a case of a patient with stage IV melanoma who exhibited a clinical response to ipilimumab treatment. Cancer exome–guided analysis of T-cell reactivity in this patient revealed reactivity against two neoantigens, including a dominant T-cell response against a mutant epitope of the ATR (ataxia telangiectasia and Rad3 related) gene product that increased strongly after ipilimumab treatment. These data provide the first demonstration (to our knowledge) of cancer exome–guided analysis to dissect the effects of melanoma immunotherapy.


Science Translational Medicine | 2014

Anti–CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response

Pia Kvistborg; Daisy Philips; Sander Kelderman; Lois Hageman; Christian Ottensmeier; Deborah Joseph-Pietras; Marij J. P. Welters; Sjoerd H. van der Burg; Ellen Kapiteijn; Olivier Michielin; Emanuela Romano; Carsten Linnemann; Daniel E. Speiser; Christian U. Blank; John B. A. G. Haanen; Ton N. M. Schumacher

Anti–CTLA-4 treatment increases the diversity of the melanoma-specific CD8 T cell response. Anti–CTLA-4 Therapy Expands T Cell Range An antibody to the immune inhibitory molecule CTLA-4, ipilimumab, can improve survival in patients with advanced melanoma. However, how anti–CTLA-4 works to improve the tumor immune response in humans remains unclear. Now, Kvistborg et al. show that although the magnitude of T cell responses was largely unaltered after therapy, the number of different T cell responses was significantly increased. Indeed, this increased breadth suggests that anti–CTLA-4 may work by increasing priming of T cells to tumor-related antigens rather than boosting preexisting immune responses. If so, other strategies that improve the range of T cells may have similar success battling cancer. Anti–CTLA-4 treatment improves the survival of patients with advanced-stage melanoma. However, although the anti–CTLA-4 antibody ipilimumab is now an approved treatment for patients with metastatic disease, it remains unknown by which mechanism it boosts tumor-specific T cell activity. In particular, it is unclear whether treatment amplifies previously induced T cell responses or whether it induces new tumor-specific T cell reactivities. Using a combination ultraviolet (UV)–induced peptide exchange and peptide–major histocompatibility complex (pMHC) combinatorial coding, we monitored immune reactivity against a panel of 145 melanoma-associated epitopes in a cohort of patients receiving anti–CTLA-4 treatment. Comparison of pre- and posttreatment T cell reactivities in peripheral blood mononuclear cell samples of 40 melanoma patients demonstrated that anti–CTLA-4 treatment induces a significant increase in the number of detectable melanoma-specific CD8 T cell responses (P = 0.0009). In striking contrast, the magnitude of both virus-specific and melanoma-specific T cell responses that were already detected before start of therapy remained unaltered by treatment (P = 0.74). The observation that anti–CTLA-4 treatment induces a significant number of newly detected T cell responses—but only infrequently boosts preexisting immune responses—provides strong evidence for anti–CTLA-4 therapy–enhanced T cell priming as a component of the clinical mode of action.


The EMBO Journal | 2012

The cancer antigenome

Bianca Heemskerk; Pia Kvistborg; Ton N. M. Schumacher

Cancer cells deviate from normal body cells in two immunologically important ways. First, tumour cells carry tens to hundreds of protein‐changing mutations that are either responsible for cellular transformation or that have accumulated as mere passengers. Second, as a consequence of genetic and epigenetic alterations, tumour cells express a series of proteins that are normally not present or present at lower levels. These changes lead to the presentation of an altered repertoire of MHC class I‐associated peptides. Importantly, while there is now strong clinical evidence that cytotoxic T‐cell activity against such tumour‐associated antigens can lead to cancer regression, at present we fail to understand which tumour‐associated antigens form the prime targets in effective immunotherapies. Here, we describe how recent developments in cancer genomics will make it feasible to establish the repertoire of tumour‐associated epitopes on a patient‐specific basis. The elucidation of this ‘cancer antigenome’ will be valuable to reveal how clinically successful immunotherapies mediate their effect. Furthermore, the description of the cancer antigenome should form the basis of novel forms of personalized cancer immunotherapy.


OncoImmunology | 2012

TIL therapy broadens the tumor-reactive CD8+ T cell compartment in melanoma patients

Pia Kvistborg; Chengyi Jenny Shu; Bianca Heemskerk; Manuel Fankhauser; Charlotte Albæk Thrue; Mireille Toebes; Nienke van Rooij; Carsten Linnemann; Marit M. van Buuren; Jos Urbanus; Joost B. Beltman; Per thor Straten; Yong F. Li; Paul F. Robbins; Michal J. Besser; Jacob Schachter; Gemma G. Kenter; Mark E. Dudley; Steven A. Rosenberg; John B. A. G. Haanen; Sine Reker Hadrup; Ton N. M. Schumacher

There is strong evidence that both adoptive T cell transfer and T cell checkpoint blockade can lead to regression of human melanoma. However, little data are available on the effect of these cancer therapies on the tumor-reactive T cell compartment. To address this issue we have profiled therapy-induced T cell reactivity against a panel of 145 melanoma-associated CD8+ T cell epitopes. Using this approach, we demonstrate that individual tumor-infiltrating lymphocyte cell products from melanoma patients contain unique patterns of reactivity against shared melanoma-associated antigens, and that the combined magnitude of these responses is surprisingly low. Importantly, TIL therapy increases the breadth of the tumor-reactive T cell compartment in vivo, and T cell reactivity observed post-therapy can almost in full be explained by the reactivity observed within the matched cell product. These results establish the value of high-throughput monitoring for the analysis of immuno-active therapeutics and suggest that the clinical efficacy of TIL therapy can be enhanced by the preparation of more defined tumor-reactive T cell products.


Nature Medicine | 2013

High-throughput identification of antigen-specific TCRs by TCR gene capture

Carsten Linnemann; Bianca Heemskerk; Pia Kvistborg; Roelof Jc Kluin; Dmitriy A. Bolotin; Xiaojing Chen; Kaspar Bresser; Marja Nieuwland; Remko Schotte; Samira Michels; Lorenz Jahn; Pleun Hombrink; Nicolas Legrand; Chengyi Jenny Shu; Ilgar Z. Mamedov; Arno Velds; Christian U. Blank; John B. A. G. Haanen; Maria A. Turchaninova; Ron M. Kerkhoven; Hergen Spits; Sine Reker Hadrup; Mirjam H.M. Heemskerk; Thomas Blankenstein; Dmitriy M. Chudakov; Gavin M. Bendle; Ton N. M. Schumacher

The transfer of T cell receptor (TCR) genes into patient T cells is a promising approach for the treatment of both viral infections and cancer. Although efficient methods exist to identify antibodies for the treatment of these diseases, comparable strategies to identify TCRs have been lacking. We have developed a high-throughput DNA-based strategy to identify TCR sequences by the capture and sequencing of genomic DNA fragments encoding the TCR genes. We establish the value of this approach by assembling a large library of cancer germline tumor antigen–reactive TCRs. Furthermore, by exploiting the quantitative nature of TCR gene capture, we show the feasibility of identifying antigen-specific TCRs in oligoclonal T cell populations from either human material or TCR-humanized mice. Finally, we demonstrate the ability to identify tumor-reactive TCRs within intratumoral T cell subsets without knowledge of antigen specificities, which may be the first step toward the development of autologous TCR gene therapy to target patient-specific neoantigens in human cancer.


Nature Protocols | 2012

Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers

Rikke Andersen; Pia Kvistborg; Thomas Mørch Frøsig; Natasja Wulff Pedersen; Rikke Birgitte Lyngaa; Arnold H. Bakker; Chengyi Jenny Shu; Per thor Straten; Ton N. M. Schumacher; Sine Reker Hadrup

Fluorescently labeled multimeric complexes of peptide-MHC, the molecular entities recognized by the T cell receptor, have become essential reagents for detection of antigen-specific CD8+ T cells by flow cytometry. Here we present a method for high-throughput parallel detection of antigen-specific T cells by combinatorial encoding of MHC multimers. Peptide-MHC complexes are produced by UV-mediated MHC peptide exchange and multimerized in the form of streptavidin-fluorochrome conjugates. Eight different fluorochromes are used for the generation of MHC multimers and, by a two-dimensional combinatorial matrix, these eight fluorochromes are combined to generate 28 unique two-color codes. By the use of combinatorial encoding, a large number of different T cell populations can be detected in a single sample. The method can be used for T cell epitope mapping, and also for the monitoring of CD8+ immune responses during cancer and infectious disease or after immunotherapy. One panel of 28 combinatorially encoded MHC multimers can be prepared in 4 h. Staining and detection takes a further 3 h.


Journal of Clinical Oncology | 2016

Phase II Study of Autologous Monocyte-Derived mRNA Electroporated Dendritic Cells (TriMixDC-MEL) Plus Ipilimumab in Patients With Pretreated Advanced Melanoma

Sofie Wilgenhof; Jurgen Corthals; Carlo Heirman; Nicolas van Baren; Sophie Lucas; Pia Kvistborg; Kris Thielemans; Bart Neyns

PURPOSE Autologous monocyte-derived dendritic cells (DCs) electroporated with synthetic mRNA (TriMixDC-MEL) are immunogenic and have antitumor activity as a monotherapy in patients with pretreated advanced melanoma. Ipilimumab, an immunoglobulin G1 monoclonal antibody directed against the cytotoxic T-lymphocyte-associated protein 4 receptor that counteracts physiologic suppression of T-cell function, improves the overall survival of patients with advanced melanoma. This phase II study investigated the combination of TriMixDC-MEL and ipilimumab in patients with pretreated advanced melanoma. PATIENTS AND METHODS Thirty-nine patients were treated with TriMixDC-MEL (4 × 10(6) cells administered intradermally and 20 × 10(6) cells administered intravenously) plus ipilimumab (10 mg/kg every 3 weeks for a total of four administrations, followed by maintenance therapy every 12 weeks in patients who remained progression free). Six-month disease control rate according to the immune-related response criteria served as the primary end point. RESULTS The 6-month disease control rate was 51% (95% CI, 36% to 67%), and the overall tumor response rate was 38% (including eight complete and seven partial responses). Seven complete responses and one partial tumor response are ongoing after a median follow-up time of 36 months (range, 22 to 43 months). The most common treatment-related adverse events (all grades) consisted of local DC injection site skin reactions (100%), transient post-DC infusion chills (38%) and flu-like symptoms (84%), dermatitis (64%), hepatitis (13%), hypophysitis (15%), and diarrhea/colitis (15%). Grade 3 or 4 immune-related adverse events occurred in 36% of patients. There was no grade 5 adverse event. CONCLUSION The combination of TriMixDC-MEL and ipilimumab is tolerable and results in an encouraging rate of highly durable tumor responses in patients with pretreated advanced melanoma.


Journal of Immunology | 2005

Spontaneous Immunity against Bcl-xL in Cancer Patients

Mads Hald Andersen; Sine Reker; Pia Kvistborg; Jiirgen C. Becker; Per thor Straten

It is well-established that peptide epitopes derived from human tumor-associated Ags can be recognized by CTL in the context of the MHC molecule. However, the vast majority of Ags described are not vital for survival and growth of the tumor cells, and immunoselection of Ag-loss variants during immunotherapy has been demonstrated in several cases. Malfunctions in death pathways observed in human cancers are often due to overexpression of antiapoptotic proteins in the Bcl-2 protein family, i.e., Bcl-2, Mcl-1, and Bcl-xL. These antiapoptotic proteins are implicated in cancer development, tumor progression, and drug resistance. The general overexpression of the antiapoptotic members of the Bcl-2 family in cancer and the fact that down-regulation or loss of expression of these proteins as a means of immune escape would impair sustained tumor growth makes them very attractive targets for anticancer immunotherapy. Recently, we identified spontaneous T cell responses against Bcl-2- and Mcl-1-derived peptides in patients suffering from cancers of different origin. In this study, we demonstrate that Bcl-xL is a target for T cell recognition in cancer patients. Thus, we describe spontaneous HLA-A2-restricted cytotoxic T cell responses against peptide epitopes derived from Bcl-xL by means of ELISPOT and flow cytometry stainings, whereas no responses were detected against any of the Bcl-xL epitopes in any healthy controls. Moreover, Bcl-xL-specific T cells are cytotoxic against HLA-matched cancer cells of different origin. Thus, cellular immune responses against apoptosis inhibitors like the Bcl-2 family proteins appear to represent a general feature in cancer.


Science Translational Medicine | 2016

The Human Vaccines Project: A roadmap for cancer vaccine development

Pedro Romero; Jacques Banchereau; Nina Bhardwaj; Mark Cockett; Mary L. Disis; Glenn Dranoff; Eli Gilboa; Scott A. Hammond; Robert M. Hershberg; Alan J. Korman; Pia Kvistborg; Cornelis J. M. Melief; Ira Mellman; A. Karolina Palucka; Irina Redchenko; Harlan Robins; Federica Sallusto; Theodore Schenkelberg; Stephen P. Schoenberger; Jeffrey A. Sosman; Özlem Türeci; Benoît Van den Eynde; Wayne C. Koff; George Couko

A concerted international effort is necessary to achieve clinically effective cancer vaccines. Cancer vaccine development has been vigorously pursued for 40 years. Immunity to tumor antigens can be elicited by most vaccines tested, but their clinical efficacy remains modest. We argue that a concerted international effort is necessary to understand the human antitumor immune response and achieve clinically effective cancer vaccines.

Collaboration


Dive into the Pia Kvistborg's collaboration.

Top Co-Authors

Avatar

Ton N. M. Schumacher

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

John B. A. G. Haanen

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Christian U. Blank

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bianca Heemskerk

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Sine Reker Hadrup

Technical University of Denmark

View shared research outputs
Top Co-Authors

Avatar

Marit M. van Buuren

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Mads Hald Andersen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Daisy Philips

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Nienke van Rooij

Netherlands Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge