Network


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

Hotspot


Dive into the research topics where Laetitia E. Lamberts is active.

Publication


Featured researches published by Laetitia E. Lamberts.


Hepatology | 2011

Three ulcerative colitis susceptibility loci are associated with primary sclerosing cholangitis and indicate a role for IL2, REL and CARD9

Marcel Janse; Laetitia E. Lamberts; Lude Franke; Soumya Raychaudhuri; Eva Ellinghaus; Kirsten Muri Boberg; Espen Melum; Trine Folseraas; Erik Schrumpf; Annika Bergquist; Einar Björnsson; Jingyuan Fu; Harm-Jan Westra; Harry J.M. Groen; Rudolf S. N. Fehrmann; Joanna Smolonska; Leonard H. van den Berg; Roel A. Ophoff; Robert J. Porte; Tobias J. Weismüller; Jochen Wedemeyer; Christoph Schramm; Martina Sterneck; Rainer Günther; Felix Braun; Severine Vermeire; Liesbet Henckaerts; Cisca Wijmenga; Cyriel Y. Ponsioen; Stefan Schreiber

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease characterized by inflammation and fibrosis of the bile ducts. Both environmental and genetic factors contribute to its pathogenesis. To further clarify its genetic background, we investigated susceptibility loci recently identified for ulcerative colitis (UC) in a large cohort of 1,186 PSC patients and 1,748 controls. Single nucleotide polymorphisms (SNPs) tagging 13 UC susceptibility loci were initially genotyped in 854 PSC patients and 1,491 controls from Benelux (331 cases, 735 controls), Germany (265 cases, 368 controls), and Scandinavia (258 cases, 388 controls). Subsequently, a joint analysis was performed with an independent second Scandinavian cohort (332 cases, 257 controls). SNPs at chromosomes 2p16 (P‐value 4.12 × 10−4), 4q27 (P‐value 4.10 × 10−5), and 9q34 (P‐value 8.41 × 10−4) were associated with PSC in the joint analysis after correcting for multiple testing. In PSC patients without inflammatory bowel disease (IBD), SNPs at 4q27 and 9q34 were nominally associated (P < 0.05). We applied additional in silico analyses to identify likely candidate genes at PSC susceptibility loci. To identify nonrandom, evidence‐based links we used GRAIL (Gene Relationships Across Implicated Loci) analysis showing interconnectivity between genes in six out of in total nine PSC‐associated regions. Expression quantitative trait analysis from 1,469 Dutch and UK individuals demonstrated that five out of nine SNPs had an effect on cis‐gene expression. These analyses prioritized IL2, CARD9, and REL as novel candidates. Conclusion: We have identified three UC susceptibility loci to be associated with PSC, harboring the putative candidate genes REL, IL2, and CARD9. These results add to the scarce knowledge on the genetic background of PSC and imply an important role for both innate and adaptive immunological factors. (HEPATOLOGY 2011;)


Journal of Clinical Oncology | 2015

Antibody Positron Emission Tomography Imaging in Anticancer Drug Development

Laetitia E. Lamberts; Simon Williams; Anton G.T. Terwisscha van Scheltinga; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Jourik A. Gietema; Adrienne H. Brouwers; Elisabeth G.E. de Vries

More than 50 monoclonal antibodies (mAbs), including several antibody-drug conjugates, are in advanced clinical development, forming an important part of the many molecularly targeted anticancer therapeutics currently in development. Drug development is a relatively slow and expensive process, limiting the number of drugs that can be brought into late-stage trials. Development decisions could benefit from quantitative biomarkers, enabling visualization of the tissue distribution of (potentially modified) therapeutic mAbs to confirm effective whole-body target expression, engagement, and modulation and to evaluate heterogeneity across lesions and patients. Such biomarkers may be realized with positron emission tomography imaging of radioactively labeled antibodies, a process called immunoPET. This approach could potentially increase the power and value of early trials by improving patient selection, optimizing dose and schedule, and rationalizing observed drug responses. In this review, we summarize the available literature and the status of clinical trials regarding the potential of immunoPET during early anticancer drug development.


Pharmacology & Therapeutics | 2014

HER3, serious partner in crime Therapeutic approaches and potential biomarkers for effect of HER3-targeting

Arjan Kol; Anton G.T. Terwisscha van Scheltinga; Hetty Timmer-Bosscha; Laetitia E. Lamberts; Frederike Bensch; Elisabeth G.E. de Vries; Carolina P. Schröder

The human epidermal growth factor receptor (HER) family members are targeted by a growing numbers of small molecules and monoclonal antibodies. Resistance against the epidermal growth factor receptor (EGFR) and HER2-targeting agents is a clinically relevant problem forcing research on optimizing targeting of the HER family. In view of its overexpression in tumors, and compensatory role in HER signaling, HER3 has gained much interest as a potential additional target within the HER family. It is the only member of the HER family lacking intrinsic tyrosine kinase activity and therefore its role in cancer has long been underestimated. Drugs that block HER3 or interfere with HER3 dimer signaling, including fully human anti-HER3 antibodies, bispecific antibodies and tyrosine kinase inhibitors (TKIs), are currently becoming available. Several compounds have already entered clinical trial. In the meantime potential biomarkers are tested such as tumor analysis of HER3 expression, functional assays for downstream effector molecules and molecular imaging techniques. This review describes the biology and relevance of HER3 in cancer, agents targeting HER3 and potential biomarkers for effect of HER3-targeting.


Clinical Cancer Research | 2017

Tumor-specific uptake of fluorescent bevacizumab-IRDye800CW microdosing in patients with primary breast cancer: a phase I feasibility study

Laetitia E. Lamberts; Maximillian Koch; Johannes S. de Jong; Arthur Adams; Jürgen Glatz; Mariëtte E.G. Kranendonk; Anton G.T. Terwisscha van Scheltinga; Liesbeth Jansen; Jakob de Vries; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Annelies Jorritsma-Smit; Matthijs D. Linssen; Esther de Boer; Bert van der Vegt; Wouter B. Nagengast; Sjoerd G. Elias; Sabrina Oliveira; Arjen J. Witkamp; Willem P. Th. M. Mali; Elsken van der Wall; Paul J. van Diest; Elisabeth G.E. de Vries; Vasilis Ntziachristos; Gooitzen M. van Dam

Purpose: To provide proof of principle of safety, breast tumor–specific uptake, and positive tumor margin assessment of the systemically administered near-infrared fluorescent tracer bevacizumab–IRDye800CW targeting VEGF-A in patients with breast cancer. Experimental Design: Twenty patients with primary invasive breast cancer eligible for primary surgery received 4.5 mg bevacizumab–IRDye800CW as intravenous bolus injection. Safety aspects were assessed as well as tracer uptake and tumor delineation during surgery and ex vivo in surgical specimens using an optical imaging system. Ex vivo multiplexed histopathology analyses were performed for evaluation of biodistribution of tracer uptake and coregistration of tumor tissue and healthy tissue. Results: None of the patients experienced adverse events. Tracer levels in primary tumor tissue were higher compared with those in the tumor margin (P < 0.05) and healthy tissue (P < 0.0001). VEGF-A tumor levels also correlated with tracer levels (r = 0.63, P < 0.0002). All but one tumor showed specific tracer uptake. Two of 20 surgically excised lumps contained microscopic positive margins detected ex vivo by fluorescent macro- and microscopy and confirmed at the cellular level. Conclusions: Our study shows that systemic administration of the bevacizumab–IRDye800CW tracer is safe for breast cancer guidance and confirms tumor and tumor margin uptake as evaluated by a systematic validation methodology. The findings are a step toward a phase II dose-finding study aimed at in vivo margin assessment and point to a novel drug assessment tool that provides a detailed picture of drug distribution in the tumor tissue. Clin Cancer Res; 23(11); 2730–41. ©2016 AACR.


Pharmacology & Therapeutics | 2014

Associate editor: B. TeicherHER3, serious partner in crime: Therapeutic approaches and potential biomarkers for effect of HER3-targeting

Arjan Kol; Anton G.T. Terwisscha van Scheltinga; Hetty Timmer-Bosscha; Laetitia E. Lamberts; Frederike Bensch; Elisabeth G.E. de Vries; Carolina P. Schröder

The human epidermal growth factor receptor (HER) family members are targeted by a growing numbers of small molecules and monoclonal antibodies. Resistance against the epidermal growth factor receptor (EGFR) and HER2-targeting agents is a clinically relevant problem forcing research on optimizing targeting of the HER family. In view of its overexpression in tumors, and compensatory role in HER signaling, HER3 has gained much interest as a potential additional target within the HER family. It is the only member of the HER family lacking intrinsic tyrosine kinase activity and therefore its role in cancer has long been underestimated. Drugs that block HER3 or interfere with HER3 dimer signaling, including fully human anti-HER3 antibodies, bispecific antibodies and tyrosine kinase inhibitors (TKIs), are currently becoming available. Several compounds have already entered clinical trial. In the meantime potential biomarkers are tested such as tumor analysis of HER3 expression, functional assays for downstream effector molecules and molecular imaging techniques. This review describes the biology and relevance of HER3 in cancer, agents targeting HER3 and potential biomarkers for effect of HER3-targeting.


Digestive and Liver Disease | 2011

Immune-mediated diseases in primary sclerosing cholangitis

Laetitia E. Lamberts; Marcel Janse; Elizabeth B. Haagsma; Arie P. van den Berg; Rinse K. Weersma

BACKGROUND Primary sclerosing cholangitis is a chronic cholestatic liver disease. An immune aetiology is suggested by associations between PSC and inflammatory bowel disease. Data on concomitant prevalence of other immune-mediated diseases is limited. AIM To assess the prevalence of concomitant immune-mediated diseases and the impact on disease outcome in PSC. METHODS We included 241 patients and retrospectively reviewed medical charts. RESULTS Altogether 172 (71.4%) patients had concomitant immune-mediated disease, including IBD (149, 61.8%), autoimmune hepatitis (15, 6.2%) and other immune-mediated diseases (47, 19.5%). Thirty nine patients (22.7%) had more than one immune-mediated disease other than PSC. Most frequent extrahepatic non-IBD immune-mediated diseases were sarcoidosis, thyroid disease, and type I diabetes mellitus. Age at PSC diagnosis was lower in patients with IBD. In patients with other immune-mediated diseases than autoimmune hepatitis or IBD, age at PSC diagnosis was higher. Younger age at diagnosis and concomitant IBD related to longer survival till death or liver transplantation. CONCLUSIONS In a large PSC population, a high prevalence of concomitant immune-mediated diseases was found. IBD occurred more often in early-acquired PSC, and the other immune-mediated diseases more often in later-acquired PSC. No effect on outcome was found for non-IBD immune mediated disease.


Molecular Cancer Therapeutics | 2016

Phase I Study of DMOT4039A, an Antibody-Drug Conjugate Targeting Mesothelin, in Patients with Unresectable Pancreatic or Platinum-Resistant Ovarian Cancer

Colin D. Weekes; Laetitia E. Lamberts; Mitesh J. Borad; Johannes Voortman; Robert R. McWilliams; Jennifer R. Diamond; Elisabeth G.E. de Vries; Henk M.W. Verheul; Christopher Hanyoung Lieu; George P. Kim; Yulei Wang; Suzie J. Scales; Divya Samineni; Flavia Brunstein; YounJeong Choi; Daniel J. Maslyar; Gerardo Colon-Otero

DMOT4039A, a humanized anti-mesothelin mAb conjugated to the antimitotic agent monomethyl auristatin E (MMAE), was given to patients with pancreatic and ovarian cancer every 3 weeks (0.2–2.8 mg/kg; q3w) or weekly (0.8–1.2 mg/kg). A 3+3 design was used for dose escalation followed by expansion at the recommended phase II dose (RP2D) to evaluate safety and pharmacokinetics. Antitumor response was evaluated per RECIST 1.1 and serum CA19-9 or CA125 declines. Tumor mesothelin expression was determined by IHC. Seventy-one patients (40 pancreatic cancer; 31 ovarian cancer) were treated with DMOT4039A. For the q3w schedule (n = 54), the MTD and RP2D was 2.4 mg/kg, with dose-limiting toxicities of grade 3 hyperglycemia and grade 3 hypophosphatemia at 2.8 mg/kg. For the weekly schedule (n = 17), the maximum assessed dose was 1.2 mg/kg, with further dose escalations deferred because of toxicities limiting scheduled retreatment in later cycles, and therefore the RP2D level for the weekly regimen was determined to be 1 mg/kg. Across both schedules, the most common toxicities were gastrointestinal and constitutional. Treatment-related serious adverse events occurred in 6 patients; 4 patients continued treatment following dose reductions. Drug exposure as measured by antibody-conjugated MMAE and total antibody was generally dose proportional over all dose levels on both schedules. A total of 6 patients had confirmed partial responses (4 ovarian; 2 pancreatic) with DMOT4039A at 2.4 to 2.8 mg/kg i.v. q3w. DMOT4039A administered at doses up to 2.4 mg/kg q3w and 1.0 mg/kg weekly has a tolerable safety profile and antitumor activity in both pancreatic and ovarian cancer. Mol Cancer Ther; 15(3); 439–47. ©2016 AACR.


Cancer Research | 2017

Threshold Analysis and Biodistribution of Fluorescently Labeled Bevacizumab in Human Breast Cancer.

Maximilian Koch; Johannes S. de Jong; Jürgen Glatz; Panagiotis Symvoulidis; Laetitia E. Lamberts; Arthur Adams; Mariëtte E.G. Kranendonk; Anton G.T. Terwisscha van Scheltinga; Michaela Aichler; Liesbeth Jansen; Jakob de Vries; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Annelies Jorritsma-Smit; Matthijs D. Linssen; Esther de Boer; Bert van der Vegt; Wouter B. Nagengast; Sjoerd G. Elias; Sabrina Oliveira; Arjen J. Witkamp; Willem P. Th. M. Mali; Elsken van der Wall; P. Beatriz Garcia-Allende; Paul J. van Diest; Elisabeth G.E. de Vries; Axel Walch; Gooitzen M. van Dam; Vasilis Ntziachristos

In vivo tumor labeling with fluorescent agents may assist endoscopic and surgical guidance for cancer therapy as well as create opportunities to directly observe cancer biology in patients. However, malignant and nonmalignant tissues are usually distinguished on fluorescence images by applying empirically determined fluorescence intensity thresholds. Here, we report the development of fSTREAM, a set of analytic methods designed to streamline the analysis of surgically excised breast tissues by collecting and statistically processing hybrid multiscale fluorescence, color, and histology readouts toward precision fluorescence imaging. fSTREAM addresses core questions of how to relate fluorescence intensity to tumor tissue and how to quantitatively assign a normalized threshold that sufficiently differentiates tumor tissue from healthy tissue. Using fSTREAM we assessed human breast tumors stained in vivo with fluorescent bevacizumab at microdose levels. Showing that detection of such levels is achievable, we validated fSTREAM for high-resolution mapping of the spatial pattern of labeled antibody and its relation to the underlying cancer pathophysiology and tumor border on a per patient basis. We demonstrated a 98% sensitivity and 79% specificity when using labeled bevacizumab to outline the tumor mass. Overall, our results illustrate a quantitative approach to relate fluorescence signals to malignant tissues and improve the theranostic application of fluorescence molecular imaging. Cancer Res; 77(3); 623-31. ©2016 AACR.


Oncotarget | 2015

Functional genomic mRNA profiling of a large cancer data base demonstrates mesothelin overexpression in a broad range of tumor types

Laetitia E. Lamberts; Derk Jan De Groot; Rico D. Bense; Elisabeth G.E. de Vries; Rudolf S. N. Fehrmann

The membrane bound glycoprotein mesothelin (MSLN) is a highly specific tumor marker, which is currently exploited as target for drugs. There are only limited data available on MSLN expression by human tumors. Therefore we determined overexpression of MSLN across different tumor types with Functional Genomic mRNA (FGM) profiling of a large cancer database. Results were compared with data in articles reporting immunohistochemical (IHC) MSLN tumor expression. FGM profiling is a technique that allows prediction of biologically relevant overexpression of proteins from a robust data set of mRNA microarrays. This technique was used in a database comprising 19,746 tumors to identify for 41 tumor types the percentage of samples with an overexpression of MSLN compared to a normal background. A literature search was performed to compare the FGM profiling data with studies reporting IHC MSLN tumor expression. FGM profiling showed MSLN overexpression in gastrointestinal (12–36%) and gynecological tumors (20–66%), non-small cell lung cancer (21%) and synovial sarcomas (30%). The overexpression found in thyroid cancers (5%) and renal cell cancers (10%) was not yet reported with IHC analyses. We observed that MSLN amplification rate within esophageal cancer depends on the histotype (31% for adenocarcinomas versus 3% for squamous-cell carcinomas). Subset analysis in breast cancer showed MSLN amplification rates of 28% in triple-negative breast cancer (TNBC) and 33% in basal-like breast cancer. Further subtype analysis of TNBCs showed the highest amplification rate (42%) in the basal-like 1 subtype and the lowest amplification rate (9%) in the luminal androgen receptor subtype.


European Journal of Pharmacology | 2013

Molecular imaging for monitoring treatment response in breast cancer patients

Frederike Bensch; Michel van Kruchten; Laetitia E. Lamberts; Carolien P. Schröder; Geke A.P. Hospers; Adrienne H. Brouwers; Marcel A. T. M. van Vugt; Elisabeth G.E. de Vries

Currently, tumour response following drug treatment is based on measurement of anatomical size changes. This is often done according to Response Evaluation Criteria in Solid Tumours (RECIST) and is generally performed every 2-3 cycles. Bone metastases, being the most common site of distant metastases in breast cancer, are not measurable by RECIST. The standard response measurement provides no insight in changes of molecular characteristics. In the era of targeted medicine, knowledge of specific molecular tumour characteristics becomes more important. A potential way to assess this is by means of molecular imaging. Molecular imaging can visualise general tumour processes, such as glucose metabolism with (18)F-fluorodeoxyglucose ((18)F-FDG) and DNA synthesis with (18)F-fluorodeoxythymidine ((18)F-FLT). In addition, an increasing number of more specific targets, such as hormone receptors, growth factor receptors, and growth factors can be visualised. In the future molecular imaging may thus be of value for personalised treatment-selection by providing insight in the expression of these drug targets. Additionally, when molecular changes can be detected early during therapy, this may serve as early predictor of response. However, in order to define clinical utility of this approach results from (ongoing) clinical trials is required. In this review we summarise the potential role of molecular imaging of general tumour processes as well as hormone receptors, growth factor receptors, and tumour micro-environment for predicting and monitoring treatment response in breast cancer patients.

Collaboration


Dive into the Laetitia E. Lamberts's collaboration.

Top Co-Authors

Avatar

Elisabeth G.E. de Vries

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Carolien P. Schröder

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adrienne H. Brouwers

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Frederike Bensch

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marjolijn N. Lub-de Hooge

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Annelies Jorritsma-Smit

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jourik A. Gietema

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marcel Janse

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge