Network


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

Hotspot


Dive into the research topics where Bianca Heemskerk is active.

Publication


Featured researches published by Bianca Heemskerk.


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.


Journal of Immunology | 2006

Gene Transfer of Tumor-Reactive TCR Confers Both High Avidity and Tumor Reactivity to Nonreactive Peripheral Blood Mononuclear Cells and Tumor-Infiltrating Lymphocytes

Laura A. Johnson; Bianca Heemskerk; Daniel J. Powell; Cyrille J. Cohen; Richard A. Morgan; Mark E. Dudley; Paul F. Robbins; Steven A. Rosenberg

Cell-based antitumor immunity is driven by CD8+ cytotoxic T cells bearing TCR that recognize specific tumor-associated peptides bound to class I MHC molecules. Of several cellular proteins involved in T cell:target-cell interaction, the TCR determines specificity of binding; however, the relative amount of its contribution to cellular avidity remains unknown. To study the relationship between TCR affinity and cellular avidity, with the intent of identifying optimal TCR for gene therapy, we derived 24 MART-1:27-35 (MART-1) melanoma Ag-reactive tumor-infiltrating lymphocyte (TIL) clones from the tumors of five patients. These MART-1-reactive clones displayed a wide variety of cellular avidities. α and β TCR genes were isolated from these clones, and TCR RNA was electroporated into the same non-MART-1-reactive allogeneic donor PBMC and TIL. TCR recipient cells gained the ability to recognize both MART-1 peptide and MART-1-expressing tumors in vitro, with avidities that closely corresponded to the original TCR clones (p = 0.018–0.0003). Clone DMF5, from a TIL infusion that mediated tumor regression clinically, showed the highest avidity against MART-1 expressing tumors in vitro, both endogenously in the TIL clone, and after RNA electroporation into donor T cells. Thus, we demonstrated that the TCR appeared to be the core determinant of MART-1 Ag-specific cellular avidity in these activated T cells and that nonreactive PBMC or TIL could be made tumor-reactive with a specific and predetermined avidity. We propose that inducing expression of this highly avid TCR in patient PBMC has the potential to induce tumor regression, as an “off-the-shelf” reagent for allogeneic melanoma patient gene therapy.


Blood | 2009

Type 17 CD8+ T cells display enhanced antitumor immunity

Christian S. Hinrichs; Andrew Kaiser; Chrystal M. Paulos; Lydie Cassard; Luis Sanchez-Perez; Bianca Heemskerk; Claudia Wrzesinski; Zachary A. Borman; Pawel Muranski; Nicholas P. Restifo

Interleukin-17 (IL-17)-secreting CD8(+) T cells have been described, but they have not been thoroughly studied and they do not have a known role in cancer immunotherapy. We skewed CD8(+) T cells to secrete IL-17 through priming in Th17-polarizing conditions. IL-17-producing CD8(+) T cells demonstrated reduced expression of Eomes and diminished cytolytic differentiation in vitro. However, after adoptive transfer, these cells converted to interferon-gamma-producing effector cells and mediated regression of large, established tumors. This improved antitumor immunity was associated with increased expression of IL-7R-alpha, decreased expression of killer cell lectin-like receptor G1, and enhanced persistence of the transferred cells. This report is the first description of a cancer therapy with IL-17-secreting CD8(+) T cells. These findings have implications for the improvement of CD8(+) T cell-based adoptive immunotherapy.


Clinical Infectious Diseases | 2002

High Levels of Adenovirus DNA in Serum Correlate with Fatal Outcome of Adenovirus Infection in Children after Allogeneic Stem-Cell Transplantation

Marco W. Schilham; Eric C. J. Claas; Wouter van Zaane; Bianca Heemskerk; Jaak M. Vossen; Arjan C. Lankester; René E. M. Toes; Marcela Echavarria; Aloys C. M. Kroes; Maarten J. D. van Tol

An increase in the incidence of adenovirus (AdV) infection leading to death among children who have undergone allogeneic stem-cell transplantation has made it necessary to find new ways to monitor AdV infection. In this retrospective study, levels of AdV DNA in serum samples obtained from 36 transplant recipients with stool cultures positive for AdV were measured by polymerase chain reaction (PCR) semiquantitatively by analyzing serial dilutions of the DNA template. Six (86%) of 7 children who died of AdV infection, compared with only 2 (7%) of 29 other patients, had high serum levels of AdV DNA (detectable by PCR at a > or =100-fold dilution of the DNA template; P<.0001). High serum levels of AdV DNA were reached a mean of 18 days before death (range, 6-29 days). Quantification of adenoviral DNA in serum may prove to be a valuable tool to diagnose and monitor AdV infection and disease in immunocompromised children.


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.


The Journal of Infectious Diseases | 2005

Immune Reconstitution and Clearance of Human Adenovirus Viremia in Pediatric Stem-Cell Recipients

Bianca Heemskerk; Arjan C. Lankester; Tamara van Vreeswijk; Matthias F. C. Beersma; Eric C. J. Claas; Louise A. Veltrop-Duits; Aloys C. M. Kroes; Jaak M. Vossen; Marco W. Schilham; Maarten J. D. van Tol

BACKGROUND Human adenovirus (HAdV) infections are increasingly frequent complications of allogeneic stem-cell transplantation (SCT), especially in children. Only a few data on the correlation between immune recovery and the course of HAdV infection are available, and data on HAdV-specific responses are lacking. METHODS In a prospective study, we determined the correlation between the HAdV DNA load in plasma and lymphocyte reconstitution in 48 children after allogeneic SCT. Additionally, HAdV-specific humoral and cellular immune responses were investigated. RESULTS HAdV infection occurred in 21 patients (44%), and, in 6 of these patients, the infection progressed to viremia, as demonstrated by the presence of HAdV DNA in plasma. Low lymphocyte counts at the onset of infection were predictive of HAdV viremia. Survival of patients with HAdV viremia was associated with an increase in lymphocyte counts during the first weeks after infection. In these patients, HAdV-specific CD4+ T cell responses, as well as increases in titers of neutralizing antibody, were detected after clearance of HAdV DNA from plasma. CONCLUSIONS Lymphocyte reconstitution appears to play a crucial role in clearance of HAdV viremia and survival of the host, warranting further development of therapeutic interventions aimed at improving immune recovery.


Blood | 2008

FOXP3 expression accurately defines the population of intratumoral regulatory T cells that selectively accumulate in metastatic melanoma lesions.

Mojgan Ahmadzadeh; Aloisio Felipe-Silva; Bianca Heemskerk; Daniel J. Powell; John R. Wunderlich; Maria J. Merino; Steven A. Rosenberg

Regulatory T (T(reg)) cells are often found in human tumors; however, their functional characteristics have been difficult to evaluate due to low cell numbers and the inability to adequately distinguish between activated and T(reg) cell populations. Using a novel approach, we examined the intracellular cytokine production capacity of tumor-infiltrating T cells in the single-cell suspensions of enzymatically digested tumors to differentiate T(reg) cells from effector T cells. Similar to T(reg) cells in the peripheral blood of healthy individuals, tumor-infiltrating FOXP3(+)CD4 T cells, unlike FOXP3(-) T cells, were unable to produce IL-2 and IFN-gamma upon ex vivo stimulation, indicating that FOXP3 expression is a valid biological marker for human T(reg) cells even in the tumor microenvironment. Accordingly, we enumerated FOXP3(+)CD4 T(reg) cells in intratumoral and peritumoral sections of metastatic melanoma tumors and found a significant increase in proportion of FOXP3(+)CD4 T(reg) cells in the intratumoral compared with peritumoral areas. Moreover, their frequencies were 3- to 5-fold higher in tumors than in peripheral blood from the same patients or healthy donors, respectively. These findings demonstrate that the tumor-infiltrating CD4 T(reg) cell population is accurately depicted by FOXP3 expression, they selectively accumulate in tumors, and their frequency in peripheral blood does not properly reflect tumor microenvironment.


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.


Journal of Virology | 2003

Extensive Cross-Reactivity of CD4+ Adenovirus-Specific T Cells: Implications for Immunotherapy and Gene Therapy

Bianca Heemskerk; Louise A. Veltrop-Duits; Tamara van Vreeswijk; Monique M. ten Dam; Sebastiaan Heidt; René E. M. Toes; Maarten J. D. van Tol; Marco W. Schilham

ABSTRACT Adenovirus (Ad)-specific T-cell responses in healthy adult donors were investigated. Ad5, inactivated by methylene blue plus visible light, induced proliferation and gamma interferon (IFN-γ) production in peripheral blood mononuclear cells of the majority of donors. Responding T cells were CD4+ and produced IFN-γ upon restimulation with infectious Ad5 and Ads of different subgroups. T-cell clones showed distinct cross-reactivity patterns recognizing Ad serotypes from either one subgroup (C), two subgroups (B and C), or three subgroups (A, B, and C). This cross-reactivity of Ad-specific T cells has relevance both for Ad-based gene therapy protocols, as well as for the feasibility of T-cell-mediated adoptive immunotherapy in recipients of an allogeneic stem cell transplantation.


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.

Collaboration


Dive into the Bianca Heemskerk'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

Pia Kvistborg

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Marco W. Schilham

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mireille Toebes

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maarten J. D. van Tol

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nienke van Rooij

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Steven A. Rosenberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Arjan C. Lankester

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge