Honors & Awards


  • Rubicon Postdoctoral Fellowship Grant, Dutch Research Council (NWO/ZonMW) (June, 2023)
  • Falling Walls 2023 Intensive Track for leading female scientists, Falling Walls Foundation (January, 2023)
  • Personal grant, Marina van Damme Fund, Issued by University Fund Wageningen (November, 2020)

Professional Education


  • Doctor of Philosophy, Unlisted School (2022)
  • Master of Science, Unlisted School (2017)
  • Bachelor of Science, Unlisted School (2014)
  • Doctor of Philosophy (PhD), Netherlands Cancer Institute, Circulating tumor DNA as a biomarker in metastatic colorectal cancer (2022)
  • Master of Science (MSc), Wageningen University, Molecular Life Sciences (2017)
  • Master of Science (MSc), Wageningen University, Molecular Nutrition and Toxicology (2017)
  • Bachelor of Science (BSc), Wageningen University, Nutrition and Health (2014)

Stanford Advisors


Lab Affiliations


All Publications


  • Metastatic Colorectal Cancer Treatment Response Evaluation by Ultra-Deep Sequencing of Cell-Free DNA and Matched White Blood Cells CLINICAL CANCER RESEARCH van 't Erve, I., Medina, J. E., Leal, A., Papp, E., Phallen, J., Adleff, V., Chiao, E., Arun, A. S., Bolhuis, K., Simmons, J. K., Karandikar, A., Valkenburg, K. C., Sausen, M., Angiuoli, S., Scharpf, R. B., Punt, C. A., Meijer, G. A., Velculescu, V. E., Fijneman, R. A. 2023; 29 (5): 899-909

    Abstract

    Circulating tumor DNA (ctDNA) has the potential to guide therapy selection and monitor treatment response in patients with metastatic cancer. However, germline and clonal hematopoiesis-associated alterations can confound identification of tumor-specific mutations in cell-free DNA (cfDNA), often requiring additional sequencing of tumor tissue. The current study assessed whether ctDNA-based treatment response monitoring could be performed in a tumor tissue-independent manner by combining ultra-deep targeted sequencing analyses of cfDNA with patient-matched white blood cell (WBC)-derived DNA.In total, 183 cfDNA and 49 WBC samples, along with 28 tissue samples, from 52 patients with metastatic colorectal cancer participating in the prospective phase III CAIRO5 clinical trial were analyzed using an ultra-deep targeted sequencing liquid biopsy assay.The combined cfDNA and WBC analysis prevented false-positives due to germline or hematopoietic variants in 40% of patients. Patient-matched tumor tissue sequencing did not provide additional information. Longitudinal analyses of ctDNA were more predictive of overall survival than standard-of-care radiological response evaluation. ctDNA mutations related to primary or acquired resistance to panitumumab were identified in 42% of patients.Accurate calling of ctDNA mutations for treatment response monitoring is feasible in a tumor tissue-independent manner by combined cfDNA and patient-matched WBC genomic DNA analysis. This tissue biopsy-independent approach simplifies sample logistics and facilitates the application of liquid biopsy ctDNA testing for evaluation of emerging therapy resistance, opening new avenues for early adaptation of treatment regimens.

    View details for DOI 10.1158/1078-0432.CCR-22-2538

    View details for Web of Science ID 000991526500001

    View details for PubMedID 36534496

    View details for PubMedCentralID PMC9975664

  • KRAS A146 Mutations Are Associated With Distinct Clinical Behavior in Patients With Colorectal Liver Metastases. JCO precision oncology van 't Erve, I., Wesdorp, N. J., Medina, J. E., Ferreira, L., Leal, A., Huiskens, J., Bolhuis, K., van Waesberghe, J. T., Swijnenburg, R. J., van den Broek, D., Velculescu, V. E., Kazemier, G., Punt, C. J., Meijer, G. A., Fijneman, R. J. 2021; 5

    Abstract

    Somatic KRAS mutations occur in approximately half of the patients with metastatic colorectal cancer (mCRC). Biologic tumor characteristics differ on the basis of the KRAS mutation variant. KRAS mutations are known to influence patient prognosis and are used as predictive biomarker for treatment decisions. This study examined clinical features of patients with mCRC with a somatic mutation in KRAS G12, G13, Q61, K117, or A146.A total of 419 patients with colorectal cancer with initially unresectable liver-limited metastases, who participated in a multicenter prospective trial, were evaluated for tumor tissue KRAS mutation status. For the subgroup of patients who carried a KRAS mutation and were treated with bevacizumab and doublet or triplet chemotherapy (N = 156), pretreatment circulating tumor DNA levels were analyzed, and total tumor volume (TTV) was quantified on the pretreatment computed tomography images.Most patients carried a KRAS G12 mutation (N = 112), followed by mutations in G13 (N = 15), A146 (N = 12), Q61 (N = 9), and K117 (N = 5). High plasma circulating tumor DNA levels were observed for patients carrying a KRAS A146 mutation versus those with a KRAS G12 mutation, with median mutant allele frequencies of 48% versus 19%, respectively. Radiologic TTV revealed this difference to be associated with a higher tumor load in patients harboring a KRAS A146 mutation (median TTV 672 cm3 [A146] v 74 cm3 [G12], P = .036). Moreover, KRAS A146 mutation carriers showed inferior overall survival compared with patients with mutations in KRAS G12 (median 10.7 v 26.4 months; hazard ratio = 2.5; P = .003).Patients with mCRC with a KRAS A146 mutation represent a distinct molecular subgroup of patients with higher tumor burden and worse clinical outcomes, who might benefit from more intensive treatments. These results highlight the importance of testing colorectal cancer for all KRAS mutations in routine clinical care.

    View details for DOI 10.1200/PO.21.00223

    View details for PubMedID 34820593

    View details for PubMedCentralID PMC8608264

  • Postoperative circulating tumour DNA is associated with pathologic response and recurrence-free survival after resection of colorectal cancer liver metastases EBIOMEDICINE Bolhuis, K., van 't Erve, I., Mijnals, C., Delis-Van Diemen, P. M., Huiskens, J., Komurcu, A., Lopez-Yurda, M., van den Broek, D., Swijnenburg, R., Meijer, G. A., Punt, C. A., Fijneman, R. A. 2021; 70: 103498

    Abstract

    Recurrence rates after resection of colorectal cancer liver metastases (CRLM) are high and correlate with worse survival. Postoperative circulating tumour DNA (ctDNA) is a promising prognostic biomarker. Focusing on patients with resected CRLM, this study aimed to evaluate the association between the detection of postoperative ctDNA, pathologic response and recurrence-free survival (RFS).Twenty-three patients were selected from an ongoing phase-3 trial who underwent resection of RAS-mutant CRLM after induction systemic treatment. CtDNA analysis was performed by droplet digital PCR using blood samples collected at baseline, before and after resection. Pathologic response of CRLM was determined via the Tumour Regression Grading system.With a median follow-up of 19.6 months, the median RFS for patients with detectable (N = 6, [26%]) and undetectable (N = 17, [74%]) postoperative ctDNA was 4.8 versus 12.1 months, respectively. Among 21 patients with available tumour tissue, pathologic response in patients with detectable compared to undetectable postoperative ctDNA was found in one of six (17%) and 15 of 15 (100%) patients, respectively (p < 0.001). In univariable Cox regression analyses both postoperative detectable ctDNA (HR = 3.3, 95%CI = 1.1-9.6, p = 0.03) and pathologic non-response (HR = 4.6, 95%CI = 1.4-15, p = 0.01) were associated with poorer RFS and were strongly correlated (r = 0.88, p < 0.001). After adjusting for clinical characteristics in pairwise multivariable analyses, postoperative ctDNA status remained associated with RFS.The detection of postoperative ctDNA after secondary resection of CRLM is a promising prognostic factor for RFS and appeared to be highly correlated with pathologic response.None.

    View details for DOI 10.1016/j.ebiom.2021.103498

    View details for Web of Science ID 000689246500007

    View details for PubMedID 34333237

    View details for PubMedCentralID PMC8340125

  • Perioperative Systemic Therapy vs Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy Alone for Resectable Colorectal Peritoneal Metastases A Phase 2 Randomized Clinical Trial JAMA SURGERY Rovers, K. P., Bakkers, C., Nienhuijs, S. W., Burger, J. A., Creemers, G. M., Thijs, A. J., Brandt-Kerkhof, A. M., Madsen, E. E., van Meerten, E., Tuynman, J. B., Kusters, M., Versteeg, K. S., Aalbers, A. J., Kok, N. M., Buffart, T. E., Wiezer, M. J., Boerma, D., Los, M., de Reuver, P. R., Bremers, A. A., Verheul, H. W., Kruijff, S., de Groot, D. A., Witkamp, A. J., van Grevenstein, W. U., Koopman, M., Nederend, J., Lahaye, M. J., Kranenburg, O., Fijneman, R. A., van 't Erve, I., Snaebjornsson, P., Hemmer, P. J., Dijkgraaf, M. W., Punt, C. A., Tanis, P. J., de Hingh, I. T., Dutch Peritoneal Oncology Grp, Dutch Colorectal Canc Grp 2021; 156 (8): 710-720

    Abstract

    To date, no randomized clinical trials have investigated perioperative systemic therapy relative to cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) alone for resectable colorectal peritoneal metastases (CPM).To assess the feasibility and safety of perioperative systemic therapy in patients with resectable CPM and the response of CPM to neoadjuvant treatment.An open-label, parallel-group phase 2 randomized clinical trial in all 9 Dutch tertiary centers for the surgical treatment of CPM enrolled participants between June 15, 2017, and January 9, 2019. Participants were patients with pathologically proven isolated resectable CPM who did not receive systemic therapy within 6 months before enrollment.Randomization to perioperative systemic therapy or CRS-HIPEC alone. Perioperative systemic therapy comprised either four 3-week neoadjuvant and adjuvant cycles of CAPOX (capecitabine and oxaliplatin), six 2-week neoadjuvant and adjuvant cycles of FOLFOX (fluorouracil, leucovorin, and oxaliplatin), or six 2-week neoadjuvant cycles of FOLFIRI (fluorouracil, leucovorin, and irinotecan) and either four 3-week adjuvant cycles of capecitabine or six 2-week adjuvant cycles of fluorouracil with leucovorin. Bevacizumab was added to the first 3 (CAPOX) or 4 (FOLFOX/FOLFIRI) neoadjuvant cycles.Proportions of macroscopic complete CRS-HIPEC and Clavien-Dindo grade 3 or higher postoperative morbidity. Key secondary outcomes were centrally assessed rates of objective radiologic and major pathologic response of CPM to neoadjuvant treatment. Analyses were done modified intention-to-treat in patients starting neoadjuvant treatment (experimental arm) or undergoing upfront surgery (control arm).In 79 patients included in the analysis (43 [54%] men; mean [SD] age, 62 [10] years), experimental (n = 37) and control (n = 42) arms did not differ significantly regarding the proportions of macroscopic complete CRS-HIPEC (33 of 37 [89%] vs 36 of 42 [86%] patients; risk ratio, 1.04; 95% CI, 0.88-1.23; P = .74) and Clavien-Dindo grade 3 or higher postoperative morbidity (8 of 37 [22%] vs 14 of 42 [33%] patients; risk ratio, 0.65; 95% CI, 0.31-1.37; P = .25). No treatment-related deaths occurred. Objective radiologic and major pathologic response rates of CPM to neoadjuvant treatment were 28% (9 of 32 evaluable patients) and 38% (13 of 34 evaluable patients), respectively.In this randomized phase 2 trial in patients diagnosed with resectable CPM, perioperative systemic therapy seemed feasible, safe, and able to induce response of CPM, justifying a phase 3 trial.ClinicalTrials.gov Identifier: NCT02758951.

    View details for DOI 10.1001/jamasurg.2021.1642

    View details for Web of Science ID 000652632700004

    View details for PubMedID 34009291

    View details for PubMedCentralID PMC8135056

  • Detection of tumor-derived cell-free DNA from colorectal cancer peritoneal metastases in plasma and peritoneal fluid JOURNAL OF PATHOLOGY CLINICAL RESEARCH Van't Erve, I., Rovers, K. P., Constantinides, A., Bolhuis, K., Wassenaar, E. E., Lurvink, R. J., Huysentruyt, C. J., Snaebjornsson, P., Boerma, D., van den Broek, D., Buffart, T. E., Lahaye, M. J., Aalbers, A. J., Kok, N. M., Meijer, G. A., Punt, C. A., Kranenburg, O., de Hingh, I. T., Fijneman, R. A. 2021; 7 (3): 203-208

    Abstract

    Tumor-derived cell-free DNA (cfDNA) is an emerging biomarker for guiding the personalized treatment of patients with metastatic colorectal cancer (CRC). While patients with CRC liver metastases (CRC-LM) have relatively high levels of plasma cfDNA, little is known about patients with CRC peritoneal metastases (CRC-PM). This study evaluated the presence of tumor-derived cfDNA in plasma and peritoneal fluid (i.e. ascites or peritoneal washing) in 20 patients with isolated CRC-PM and in the plasma of 100 patients with isolated CRC-LM. Among tumor tissue KRAS/BRAF mutation carriers, tumor-derived cfDNA was detected by droplet digital polymerase chain reaction (ddPCR) in plasma of 93% of CRC-LM and 20% of CRC-PM patients and in peritoneal fluid in all CRC-PM patients. Mutant allele fraction (MAF) and mutant copies per ml (MTc/ml) were lower in CRC-PM plasma than in CRC-LM plasma (median MAF = 0.28 versus 18.9%, p < 0.0001; median MTc/ml = 21 versus 1,758, p < 0.0001). Within patients with CRC-PM, higher cfDNA levels were observed in peritoneal fluid than in plasma (median MAF = 16.4 versus 0.28%, p = 0.0019; median MTc/ml = 305 versus 21, p = 0.0034). These data imply that tumor-derived cfDNA in plasma is a poor biomarker to monitor CRC-PM. Instead, cfDNA detection in peritoneal fluid may offer an alternative to guide CRC-PM treatment decisions.

    View details for DOI 10.1002/cjp2.207

    View details for Web of Science ID 000621904100001

    View details for PubMedID 33635598

    View details for PubMedCentralID PMC8073000

  • First-line palliative systemic therapy alternated with electrostatic pressurised intraperitoneal aerosol chemotherapy (oxaliplatin) for isolated unresectable colorectal peritoneal metastases: protocol of a multicentre, single-arm, phase II study (CRC-PIPAC-II) BMJ OPEN Lurvink, R. J., Rauwerdink, P., Rovers, K. P., Wassenaar, E. E., Deenen, M. J., Nederend, J., Huysentruyt, C. R., van 't Erve, I., Fijneman, R. A., van der Hoeven, E. J., Seldenrijk, C. A., Constantinides, A., Kranenburg, O., Los, M., Herbschleb, K. H., Thijs, A. J., Creemers, G. M., Burger, J. A., Wiezer, M. J., Nienhuijs, S. W., Boerma, D., de Hingh, I. T. 2021; 11 (3): e044811

    Abstract

    Despite its increasing use, first-line palliative systemic therapy alternated with electrostatic pressurised intraperitoneal aerosol chemotherapy with oxaliplatin (ePIPAC-OX), hereinafter referred to as first-line bidirectional therapy, has never been prospectively investigated in patients with colorectal peritoneal metastases (CPM). As a first step to address this evidence gap, the present study aims to assess the safety, feasibility, antitumour activity, patient-reported outcomes, costs and systemic pharmacokinetics of first-line bidirectional therapy in patients with isolated unresectable CPM.In this single-arm, phase II study in two Dutch tertiary referral centres, 20 patients are enrolled. Key eligibility criteria are a good performance status, pathologically proven isolated unresectable CPM, no previous palliative systemic therapy for colorectal cancer, no (neo)adjuvant systemic therapy ≤6 months prior to enrolment and no previous pressurised intraperitoneal aerosol chemotherapy (PIPAC). Patients receive three cycles of bidirectional therapy. Each cycle consists of 6 weeks first-line palliative systemic therapy at the medical oncologists' decision (CAPOX-bevacizumab, FOLFOX-bevacizumab, FOLFIRI-bevacizumab or FOLFOXIRI-bevacizumab) followed by ePIPAC-OX (92 mg/m2) with an intraoperative bolus of intravenous leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2). Study treatment ends after the third ePIPAC-OX. The primary outcome is the number of patients with-and procedures leading to-grade ≥3 adverse events (Common Terminology Criteria for Adverse Events V.5.0) up to 4 weeks after the last procedure. Key secondary outcomes include the number of bidirectional cycles in each patient, treatment-related characteristics, grade ≤2 adverse events, tumour response (histopathological, cytological, radiological, biochemical, macroscopic and ascites), patient-reported outcomes, systemic pharmacokinetics of oxaliplatin, costs, progression-free survival and overall survival.This study is approved by the Dutch competent authority, a medical ethics committee and the institutional review boards of both study centres. Results will be submitted for publication in peer-reviewed medical journals and presented to patients and healthcare professionals.NL8303.

    View details for DOI 10.1136/bmjopen-2020-044811

    View details for Web of Science ID 000636355300008

    View details for PubMedID 33785492

    View details for PubMedCentralID PMC8011718

  • Diagnostic Strategies toward Clinical Implementation of Liquid Biopsy RAS/BRAF Circulating Tumor DNA Analyses in Patients with Metastatic Colorectal Cancer JOURNAL OF MOLECULAR DIAGNOSTICS van 't Erve, I., Greuter, E. E., Bolhuis, K., Vessies, D. L., Leal, A., Vink, G. R., van den Broek, D., Velculescu, V. E., Punt, C. A., Meijer, G. A., Coupe, V. H., Fijneman, R. A. 2020; 22 (12): 1430-1437

    Abstract

    Detection of KRAS, NRAS, and BRAF mutations in tumor tissue is currently used to predict resistance to treatment with anti-epidermal growth factor receptor (EGFR) antibodies in patients with metastatic colorectal cancer (mCRC). Liquid biopsies are minimally invasive, and cell-free circulating tumor DNA (ctDNA) mutation analyses may better represent tumor heterogeneity. This study examined the incorporation of liquid biopsy RAS/BRAF ctDNA analyses into diagnostic strategies to determine mCRC patient eligibility for anti-EGFR therapy. Tumor tissue and liquid biopsies were collected from 100 mCRC patients with liver-only metastases in a multicenter prospective clinical trial. Three diagnostic strategies incorporating droplet digital PCR ctDNA analyses were compared with routine tumor tissue RAS/BRAF mutation profiling using decision tree analyses. Tissue DNA mutations in KRAS, NRAS, and BRAF were present in 54%, 0%, and 3% of mCRC patients, respectively. A 93% concordance was observed between tissue DNA and liquid biopsy ctDNA mutations. The proportion of patients with RAS/BRAF alterations increased from 57% to 60% for diagnostic strategies that combined tissue and liquid biopsy mutation analyses. Consecutive RAS/BRAF ctDNA analysis followed by tissue DNA analysis in case of a liquid biopsy-negative result appeared to be the most optimal diagnostic strategy to comprehensively determine eligibility for anti-EGFR therapy in a cost-saving manner. These results highlight the potential clinical utility of liquid biopsies for detecting primary resistance to anti-EGFR-targeted therapies.

    View details for DOI 10.1016/j.jmoldx.2020.09.002

    View details for Web of Science ID 000591511100008

    View details for PubMedID 32961317

  • Repetitive electrostatic pressurised intraperitoneal aerosol chemotherapy (ePIPAC) with oxaliplatin as a palliative monotherapy for isolated unresectable colorectal peritoneal metastases: protocol of a Dutch, multicentre, open-label, single-arm, phase II study (CRC-PIPAC) BMJ OPEN Rovers, K. P., Lurvink, R. J., Wassenaar, E. E., Kootstra, T. M., Scholten, H. J., Tajzai, R., Deenen, M. J., Nederend, J., Lahaye, M. J., Huysentruyt, C. R., van't Erve, I., Fijneman, R. A., Constantinides, A., Kranenburg, O., Los, M., Thijs, A. J., Creemers, G. M., Burger, J. A., Wiezer, M. J., Boerma, D., Nienhuijs, S. W., de Hingh, I. T. 2019; 9 (7): e030408

    Abstract

    Repetitive electrostatic pressurised intraperitoneal aerosol chemotherapy with oxaliplatin (ePIPAC-OX) is offered as a palliative treatment option for patients with isolated unresectable colorectal peritoneal metastases (PM) in several centres worldwide. However, little is known about its feasibility, safety, tolerability, efficacy, costs and pharmacokinetics in this setting. This study aims to explore these parameters in patients with isolated unresectable colorectal PM who receive repetitive ePIPAC-OX as a palliative monotherapy.This multicentre, open-label, single-arm, phase II study is performed in two Dutch tertiary referral hospitals for the surgical treatment of colorectal PM. Eligible patients are adults who have histologically or cytologically proven isolated unresectable PM of a colorectal or appendiceal carcinoma, a good performance status, adequate organ functions and no symptoms of gastrointestinal obstruction. Instead of standard palliative treatment, enrolled patients receive laparoscopy-controlled ePIPAC-OX (92 mg/m2 body surface area (BSA)) with intravenous leucovorin (20 mg/m2 BSA) and bolus 5-fluorouracil (400 mg/m2 BSA) every 6 weeks. Four weeks after each procedure, patients undergo clinical, radiological and biochemical evaluation. ePIPAC-OX is repeated until disease progression, after which standard palliative treatment is (re)considered. The primary outcome is the number of patients with major toxicity (grade ≥3 according to the Common Terminology Criteria for Adverse Events v4.0) up to 4 weeks after the last ePIPAC-OX. Secondary outcomes are the environmental safety of ePIPAC-OX, procedure-related characteristics, minor toxicity, postoperative complications, hospital stay, readmissions, quality of life, costs, pharmacokinetics of oxaliplatin, progression-free survival, overall survival, and the radiological, histopathological, cytological, biochemical and macroscopic tumour response.This study is approved by an ethics committee, the Dutch competent authority and the institutional review boards of both study centres. Results are intended for publication in peer-reviewed medical journals and for presentation to patients, healthcare professionals and other stakeholders.NCT03246321, Pre-results; ISRCTN89947480, Pre-results; NTR6603, Pre-results; EudraCT: 2017-000927-29, Pre-results.

    View details for DOI 10.1136/bmjopen-2019-030408

    View details for Web of Science ID 000485269700088

    View details for PubMedID 31352425

    View details for PubMedCentralID PMC6661551

  • Changes in Circulating Levels of 25-hydroxyvitamin D3 in Breast Cancer Patients Receiving Chemotherapy NUTRITION AND CANCER-AN INTERNATIONAL JOURNAL Kok, D. E., van den Berg, M. A., Posthuma, L., van't Erve, I., van Duijnhoven, F. B., de Roos, W. K., Grosfeld, S., Los, M., Sommeijer, D. W., van Laarhoven, H. M., Winkels, R. M., Kampman, E. 2019; 71 (5): 756-766

    Abstract

    Cancer treatments, toxicities and their effects on lifestyle, may impact levels of vitamin D. The aim of this study was to determine serum 25-hydroxyvitamin D3 (25(OH)D3) levels before, directly after and 6 months after chemotherapy in breast cancer patients (n = 95), and a comparison group of women (n = 52) not diagnosed with cancer. Changes in 25(OH)D3 levels over time were compared using linear mixed models adjusted for age and season of blood sampling. Before start of chemotherapy, 25(OH)D3 levels were lower in patients (estimated marginal mean 55.8 nmol/L, 95% confidence interval (95%CI) 51.2-60.4) compared to the comparison group (67.2 nmol/L, 95%CI 61.1-73.3, P = 0.003). Directly after chemotherapy, 25(OH)D3 levels were slightly decreased (-5.1 nmol/L, 95%CI -10.7-0.5, P = 0.082), but ended up higher 6 months after chemotherapy (10.9 nmol/L, 95%CI 5.5-16.4, P < 0.001) compared to pre-chemotherapy values. In women without cancer, 25(OH)D3 levels remained stable throughout the study. Use of dietary supplements did not explain recovery of 25(OH)D3 levels after chemotherapy. We reported lower 25(OH)D3 levels in breast cancer patients, which decreased during chemotherapy, but recovered to levels observed in women without cancer within 6 months after chemotherapy. Suboptimal 25(OH)D3 levels in the majority of the participants highlight the relevance of monitoring in this vulnerable population.

    View details for DOI 10.1080/01635581.2018.1559938

    View details for Web of Science ID 000467757800006

    View details for PubMedID 30661404

  • Perioperative systemic therapy and cytoreductive surgery with HIPEC versus upfront cytoreductive surgery with HIPEC alone for isolated resectable colorectal peritoneal metastases: protocol of a multicentre, open-label, parralel-group, phase II-III, randomised, superiority study (CAIRO6) BMC CANCER Rovers, K. P., Bakkers, C., Simkens, G. M., Burger, J. A., Nienhuijs, S. W., Creemers, G. M., Thijs, A. J., Brandt-Kerkhof, A. M., Madsen, E. E., Ayez, N., de Boer, N. L., van Meerten, E., Tuynman, J. B., Kusters, M., Sluiter, N. R., Verheul, H. W., van der Vliet, H. J., Wiezer, M. J., Boerma, D., Wassenaar, E. E., Los, M., Hunting, C. B., Aalbers, A. J., Kok, N. M., Kuhlmann, K. D., Boot, H., Chalabi, M., Kruijff, S., Been, L. B., van Ginkel, R. J., de Groot, D. A., Fehrmann, R. N., de Wilt, J. W., Bremers, A. A., de Reuver, P. R., Radema, S. A., Herbschleb, K. H., van Grevenstein, W. U., Witkamp, A. J., Koopman, M., Mohammad, N., van Duyn, E. B., Mastboom, W. B., Mekenkamp, L. M., Nederend, J., Lahaye, M. J., Snaebjornsson, P., Verhoef, C., van Laarhoven, H. M., Zwinderman, A. H., Bouma, J. M., Kranenburg, O., van 't Erve, I., Fijneman, R. A., Dijkgraaf, M. W., Hemmer, P. J., Punt, C. A., Tanis, P. J., de Hingh, I. T., DPOG, DCCG 2019; 19: 390

    Abstract

    Upfront cytoreductive surgery with HIPEC (CRS-HIPEC) is the standard treatment for isolated resectable colorectal peritoneal metastases (PM) in the Netherlands. This study investigates whether addition of perioperative systemic therapy to CRS-HIPEC improves oncological outcomes.This open-label, parallel-group, phase II-III, randomised, superiority study is performed in nine Dutch tertiary referral centres. Eligible patients are adults who have a good performance status, histologically or cytologically proven resectable PM of a colorectal adenocarcinoma, no systemic colorectal metastases, no systemic therapy for colorectal cancer within six months prior to enrolment, and no previous CRS-HIPEC. Eligible patients are randomised (1:1) to perioperative systemic therapy and CRS-HIPEC (experimental arm) or upfront CRS-HIPEC alone (control arm) by using central randomisation software with minimisation stratified by a peritoneal cancer index of 0-10 or 11-20, metachronous or synchronous PM, previous systemic therapy for colorectal cancer, and HIPEC with oxaliplatin or mitomycin C. At the treating physician's discretion, perioperative systemic therapy consists of either four 3-weekly neoadjuvant and adjuvant cycles of capecitabine with oxaliplatin (CAPOX), six 2-weekly neoadjuvant and adjuvant cycles of 5-fluorouracil/leucovorin with oxaliplatin (FOLFOX), or six 2-weekly neoadjuvant cycles of 5-fluorouracil/leucovorin with irinotecan (FOLFIRI) followed by four 3-weekly (capecitabine) or six 2-weekly (5-fluorouracil/leucovorin) adjuvant cycles of fluoropyrimidine monotherapy. Bevacizumab is added to the first three (CAPOX) or four (FOLFOX/FOLFIRI) neoadjuvant cycles. The first 80 patients are enrolled in a phase II study to explore the feasibility of accrual and the feasibility, safety, and tolerance of perioperative systemic therapy. If predefined criteria of feasibility and safety are met, the study continues as a phase III study with 3-year overall survival as primary endpoint. A total of 358 patients is needed to detect the hypothesised 15% increase in 3-year overall survival (control arm 50%; experimental arm 65%). Secondary endpoints are surgical characteristics, major postoperative morbidity, progression-free survival, disease-free survival, health-related quality of life, costs, major systemic therapy related toxicity, and objective radiological and histopathological response rates.This is the first randomised study that prospectively compares oncological outcomes of perioperative systemic therapy and CRS-HIPEC with upfront CRS-HIPEC alone for isolated resectable colorectal PM.Clinicaltrials.gov/ NCT02758951 , NTR/ NTR6301 , ISRCTN/ ISRCTN15977568 , EudraCT/ 2016-001865-99 .

    View details for DOI 10.1186/s12885-019-5545-0

    View details for Web of Science ID 000465873900002

    View details for PubMedID 31023318

    View details for PubMedCentralID PMC6485075