1
|
Vliek S, Hilbers FS, van Werkhoven E, Mandjes I, Kessels R, Kleiterp S, Lips EH, Mulder L, Kayembe MT, Loo CE, Russell NS, Vrancken Peeters MJTFD, Holtkamp MJ, Schot M, Baars JW, Honkoop AH, Vulink AJE, Imholz ALT, Vrijaldenhoven S, van den Berkmortel FWPJ, Meerum Terwogt JM, Schrama JG, Kuijer P, Kroep JR, van der Padt-Pruijsten A, Wesseling J, Sonke GS, Gilhuijs KGA, Jager A, Nederlof P, Linn SC. High-dose alkylating chemotherapy in BRCA-altered triple-negative breast cancer: the randomized phase III NeoTN trial. NPJ Breast Cancer 2023; 9:75. [PMID: 37689749 PMCID: PMC10492793 DOI: 10.1038/s41523-023-00580-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Exploratory analyses of high-dose alkylating chemotherapy trials have suggested that BRCA1 or BRCA2-pathway altered (BRCA-altered) breast cancer might be particularly sensitive to this type of treatment. In this study, patients with BRCA-altered tumors who had received three initial courses of dose-dense doxorubicin and cyclophosphamide (ddAC), were randomized between a fourth ddAC course followed by high-dose carboplatin-thiotepa-cyclophosphamide or conventional chemotherapy (initially ddAC only or ddAC-capecitabine/decetaxel [CD] depending on MRI response, after amendment ddAC-carboplatin/paclitaxel [CP] for everyone). The primary endpoint was the neoadjuvant response index (NRI). Secondary endpoints included recurrence-free survival (RFS) and overall survival (OS). In total, 122 patients were randomized. No difference in NRI-score distribution (p = 0.41) was found. A statistically non-significant RFS difference was found (HR 0.54; 95% CI 0.23-1.25; p = 0.15). Exploratory RFS analyses showed benefit in stage III (n = 35; HR 0.16; 95% CI 0.03-0.75), but not stage II (n = 86; HR 1.00; 95% CI 0.30-3.30) patients. For stage III, 4-year RFS was 46% (95% CI 24-87%), 71% (95% CI 48-100%) and 88% (95% CI 74-100%), for ddAC/ddAC-CD, ddAC-CP and high-dose chemotherapy, respectively. No significant differences were found between high-dose and conventional chemotherapy in stage II-III, triple-negative, BRCA-altered breast cancer patients. Further research is needed to establish if there are patients with stage III, triple negative BRCA-altered breast cancer for whom outcomes can be improved with high-dose alkylating chemotherapy or whether the current standard neoadjuvant therapy including carboplatin and an immune checkpoint inhibitor is sufficient. Trial Registration: NCT01057069.
Collapse
Affiliation(s)
- Sonja Vliek
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Florentine S Hilbers
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Erik van Werkhoven
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- HOVON Data Center, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ingrid Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rob Kessels
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sieta Kleiterp
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lennart Mulder
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mutamba T Kayembe
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Claudette E Loo
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nicola S Russell
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marie-Jeanne T F D Vrancken Peeters
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam University Medical center, Amsterdam, The Netherlands
| | - Marjo J Holtkamp
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Margaret Schot
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joke W Baars
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aafke H Honkoop
- Department of Internal Medicine, Isala Klinieken, Zwolle, The Netherlands
| | - Annelie J E Vulink
- Division of Medical Oncology, Reinier de Graaf Hospital, Delft, The Netherlands
| | - Alex L T Imholz
- Department of Internal Medicine, Deventer Ziekenhuis, Deventer, The Netherlands
| | | | | | | | - Jolanda G Schrama
- Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Philomeen Kuijer
- Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jelle Wesseling
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kenneth G A Gilhuijs
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Petra Nederlof
- Department of Molecular diagnostics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sabine C Linn
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
2
|
Verbeek JGE, de Jong VMT, Wijnja HM, Jager A, Linn SC, Retèl VP, van Harten WH. High-dose chemotherapy with stem cell rescue to treat stage III homologous deficient breast cancer: factors influencing clinical implementation. BMC Cancer 2023; 23:26. [PMID: 36611165 PMCID: PMC9824989 DOI: 10.1186/s12885-022-10412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 12/05/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND High-dose chemotherapy with autologous stem cell rescue (HDCT) is a promising treatment for patients with stage III, HER2-negative, homologous recombination deficient (HRD) breast cancer. Clinical effectiveness and cost-effectiveness are currently under investigation in an international multicenter randomized controlled trial. To increase the chance of successful introduction of HDCT into daily clinical practice, we aimed to identify relevant factors for smooth implementation using an early comprehensive assessment framework. METHODS This is a qualitative, multi-stakeholder, exploratory research using semi-structured interviews guided by the Constructive Technology Assessment model, which evaluates the quality of a novel health technology by clinical, economic, patient-related, and organizational factors. Stakeholders were recruited by purposeful stratified sampling and interviewed until sufficient content saturation was reached. Two researchers independently created themes, categories, and subcategories by following inductive coding steps, these were verified by a third researcher. RESULTS We interviewed 28 stakeholders between June 2019 and April 2021. In total, five overarching themes and seventeen categories were identified. Important findings for optimal implementation included the structural identification and referral of all eligible patients, early integration of supportive care, multidisciplinary collaboration between- and within hospitals, (de)centralization of treatment aspects, the provision of information for patients and healthcare professionals, and compliance to new regulation for the BRCA1-like test. CONCLUSIONS In anticipation of a positive reimbursement decision, we recommend to take the highlighted implementation factors into consideration. This might expedite and guide high-quality equitable access to HDCT for patients with stage III, HER2-negative, HRD breast cancer in the Netherlands.
Collapse
Affiliation(s)
- Joost G. E. Verbeek
- grid.430814.a0000 0001 0674 1393Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands ,grid.6214.10000 0004 0399 8953Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Vincent M. T. de Jong
- grid.430814.a0000 0001 0674 1393Department of Molecular Pathology, Antoni Van Leeuwenhoek Hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hanna M. Wijnja
- grid.430814.a0000 0001 0674 1393Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Agnes Jager
- grid.508717.c0000 0004 0637 3764Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sabine C. Linn
- grid.430814.a0000 0001 0674 1393Department of Molecular Pathology, Antoni Van Leeuwenhoek Hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.430814.a0000 0001 0674 1393Department of Medical Oncology, Antoni Van Leeuwenhoek Hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.7692.a0000000090126352Department of Pathology, Utrecht University Medical Centre, Utrecht, The Netherlands
| | - Valesca P. Retèl
- grid.430814.a0000 0001 0674 1393Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands ,grid.6214.10000 0004 0399 8953Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Wim H. van Harten
- grid.430814.a0000 0001 0674 1393Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands ,grid.6214.10000 0004 0399 8953Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| |
Collapse
|
3
|
Joy AA, Vos LJ, Pituskin E, Cook SF, Bies RR, Vlahadamis A, King K, Basi SK, Meza-Junco J, Mackey JR, Stanislaus A, Damaraju VL, Damaraju S, Sawyer MB. Uridine Glucuronosyltransferase 2B7 Polymorphism-Based Pharmacogenetic Dosing of Epirubicin in FEC Chemotherapy for Early-Stage Breast Cancer. Clin Breast Cancer 2021; 21:e584-e593. [PMID: 33832852 DOI: 10.1016/j.clbc.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Epirubicin is metabolized by uridine glucuronosyltransferase 2B7 (UGT2B7). Patients homozygous for the minor allele (CC) in the UGT2B7 -161 promoter polymorphism have lower clearance and significantly higher rates of leukopenia compared to wild-type homozygote (TT) or heterozygote (CT) patients. This study was designed to determine if TT and CT genotype patients could tolerate a higher epirubicin dose compared to CC genotype patients. PATIENTS AND METHODS We studied women with histologically confirmed non-metastatic, invasive breast cancer who were scheduled to receive at least three cycles of FE100C in the (neo)adjuvant setting. Patients received standard-dose FE100C during the first 21-day cycle. Based on genotype, the epirubicin dose was escalated in the second and third cycles to 115 and 130 mg/m2 or to 120 and 140 mg/m2 for CT and TT genotype patients, respectively. The main outcome measurements were myelosuppression and dose-limiting toxicity. These were analyzed for relationships with the three genotypes. RESULTS Forty-five patients were enrolled (10 CC, 21 CT, and 14 TT genotypes) and received 100 mg/m2 of epirubicin in the first cycle. Twelve and 10 TT patients were dose escalated at the second and third cycles, respectively; 16 CT patients were dose escalated at the second and third cycles. Leukopenia, but not febrile neutropenia, was genotype and dose dependent and increased in patients with CT and TT genotypes as their dose was increased. However, the third-cycle leukopenia rates were comparable to patients with the CC genotype receiving standard-dose epirubicin. CONCLUSION Pharmacogenetically guided epirubicin dosing is well tolerated and allowed dose escalation without increased toxicity.
Collapse
Affiliation(s)
- Anil A Joy
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Larissa J Vos
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Edith Pituskin
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
| | - Sarah F Cook
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Robert R Bies
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Ann Vlahadamis
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Karen King
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sanraj K Basi
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Judith Meza-Junco
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - John R Mackey
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Avalyn Stanislaus
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vijaya L Damaraju
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sambasivarao Damaraju
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Michael B Sawyer
- Division of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada; Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Steenbruggen TG, Steggink LC, Seynaeve CM, van der Hoeven JJM, Hooning MJ, Jager A, Konings IR, Kroep JR, Smit WM, Tjan-Heijnen VCG, van der Wall E, Bins AD, Linn SC, Schaapveld M, Jacobse JN, van Leeuwen FE, Schröder CP, van Tinteren H, de Vries EGE, Sonke GS, Gietema JA. High-Dose Chemotherapy With Hematopoietic Stem Cell Transplant in Patients With High-Risk Breast Cancer and 4 or More Involved Axillary Lymph Nodes: 20-Year Follow-up of a Phase 3 Randomized Clinical Trial. JAMA Oncol 2020; 6:528-534. [PMID: 31999296 DOI: 10.1001/jamaoncol.2019.6276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Trials of adjuvant high-dose chemotherapy (HDCT) have failed to show a survival benefit in unselected patients with breast cancer, but long-term follow-up is lacking. Objective To determine 20-year efficacy and safety outcomes of a large trial of adjuvant HDCT vs conventional-dose chemotherapy (CDCT) for patients with stage III breast cancer. Design, Setting, and Participants This secondary analysis used data from a randomized phase 3 multicenter clinical trial of 885 women younger than 56 years with breast cancer and 4 or more involved axillary lymph nodes conducted from August 1, 1993, to July 31, 1999. Additional follow-up data were collected between June 1, 2016, and December 31, 2017, from medical records, general practitioners, the Dutch national statistical office, and nationwide cancer registries. Analysis was performed on an intention-to-treat basis. Statistical analysis was performed from February 1, 2018, to October 14, 2019. Interventions Participants were randomized 1:1 to receive 5 cycles of CDCT consisting of fluorouracil, 500 mg/m2, epirubicin, 90 mg/m2, and cyclophosphamide, 500 mg/m2, or HDCT in which the first 4 cycles were identical to CDCT and the fifth cycle was replaced by cyclophosphamide, 6000 mg/m2, thiotepa, 480 mg/m2, and carboplatin, 1600 mg/m2, followed by hematopoietic stem cell transplant. Main Outcomes and Measures Main end points were overall survival and safety and cumulative incidence risk of a second malignant neoplasm or cardiovascular events. Results Of the 885 women in the study (mean [SD] age, 44.5 [6.6] years), 442 were randomized to receive HDCT, and 443 were randomized to receive CDCT. With 20.4 years median follow-up (interquartile range, 19.2-22.0 years), the 20-year overall survival was 45.3% with HDCT and 41.5% with CDCT (hazard ratio, 0.89; 95% CI, 0.75-1.06). The absolute improvement in 20-year overall survival was 14.6% (hazard ratio, 0.72; 95% CI, 0.54-0.95) for patients with 10 or more invoved axillary lymph nodes and 15.4% (hazard ratio, 0.67; 95% CI, 0.42-1.05) for patients with triple-negative breast cancer. The cumulative incidence risk of a second malignant neoplasm at 20 years or major cardiovascular events was similar in both treatment groups (20-year cumulative incidence risk for second malignant neoplasm was 12.1% in the HDCT group vs 16.2% in the CDCT group, P = .10), although patients in the HDCT group more often had hypertension (21.7% vs 14.3%, P = .02), hypercholesterolemia (15.7% vs 10.6%, P = .04), and dysrhythmias (8.6% vs 4.6%, P = .005). Conclusions and Relevance High-dose chemotherapy provided no long-term survival benefit in unselected patients with stage III breast cancer but did provide improved overall survival in very high-risk patients (ie, with ≥10 involved axillary lymph nodes). High-dose chemotherapy did not affect long-term risk of a second malignant neoplasm or major cardiovascular events. Trial Registration ClinicalTrials.gov Identifier: NCT03087409.
Collapse
Affiliation(s)
- Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Lars C Steggink
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Caroline M Seynaeve
- Department of Medical Oncology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Inge R Konings
- Department of Medical Oncology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Wim M Smit
- Department of Internal Medicine/Medical Oncology, Medisch Spectrum Twente, Enschede, the Netherlands
| | | | - Elsken van der Wall
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam UMC, location AMC, Amsterdam, the Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Michael Schaapveld
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Judy N Jacobse
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Flora E van Leeuwen
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Harm van Tinteren
- Department of Biostatistics, the Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
5
|
Lips EH, Benard-Slagter A, Opdam M, Scheerman CE, Wesseling J, Hogervorst FBL, Linn SC, Savola S, Nederlof PM. BRCAness digitalMLPA profiling predicts benefit of intensified platinum-based chemotherapy in triple-negative and luminal-type breast cancer. Breast Cancer Res 2020; 22:79. [PMID: 32711554 PMCID: PMC7382055 DOI: 10.1186/s13058-020-01313-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We previously showed that BRCA-like profiles can be used to preselect individuals with the highest risk of carrying BRCA mutations but could also indicate which patients would benefit from double-strand break inducing chemotherapy. A simple, robust, and reliable assay for clinical use that utilizes limited amounts of formalin-fixed, paraffin-embedded tumor tissue to assess BRCAness status in both ER-positive and ER-negative breast cancer (BC) is currently lacking. METHODS A digital multiplex ligation-dependent probe amplification (digitalMLPA) assay was designed to detect copy number alterations required for the classification of BRCA1-like and BRCA2-like BC. The BRCA1-like classifier was trained on 71 tumors, enriched for triple-negative BC; the BRCA2-like classifier was trained on 55 tumors, enriched for luminal-type BC. A shrunken centroid-based classifier was developed and applied on an independent validation cohort. A total of 114 cases of a randomized controlled trial were analyzed, and the association of the classifier result with intensified platinum-based chemotherapy response was assessed. RESULTS The digitalMLPA BRCA1-like classifier correctly classified 91% of the BRCA1-like samples and 82% of the BRCA2-like samples. Patients with a BRCA-like tumor derived significant benefit of high-dose chemotherapy (adjusted hazard ratio (HR) 0.12, 95% CI 0.04-0.44) which was not observed in non-BRCA-like patients (HR 0.9, 95% CI 0.37-2.18) (p = 0.01). Analysis stratified for ER status showed borderline significance. CONCLUSIONS The digitalMLPA is a reliable method to detect a BRCA1- and BRCA2-like pattern on clinical samples and predicts platinum-based chemotherapy benefit in both triple-negative and luminal-type BC.
Collapse
Affiliation(s)
- Esther H Lips
- Department of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | | | - Mark Opdam
- Department of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Caroline E Scheerman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Department of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans B L Hogervorst
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sabine C Linn
- Department of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Suvi Savola
- Department of Oncogenetics, MRC Holland, Amsterdam, The Netherlands
| | - Petra M Nederlof
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
6
|
BCMA-targeting Bispecific Antibody That Simultaneously Stimulates NKG2D-enhanced Efficacy Against Multiple Myeloma. J Immunother 2020; 43:175-188. [DOI: 10.1097/cji.0000000000000320] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
7
|
Criscitiello C, Curigliano G. Tumour infiltrating lymphocytes and correlation with response to intensified platinum-based chemotherapy in BRCA-like tumours. Eur J Cancer 2020; 127:236-239. [DOI: 10.1016/j.ejca.2019.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/24/2023]
|
8
|
de Boo L, Cimino-Mathews A, Lubeck Y, Daletzakis A, Opdam M, Sanders J, Hooijberg E, van Rossum A, Loncova Z, Rieder D, Trajanoski Z, Vollebergh M, Sobral-Leite M, van de Vijver K, Broeks A, van der Wiel R, van Tinteren H, Linn S, Horlings HM, Kok M. Tumour-infiltrating lymphocytes (TILs) and BRCA-like status in stage III breast cancer patients randomised to adjuvant intensified platinum-based chemotherapy versus conventional chemotherapy. Eur J Cancer 2020; 127:240-250. [PMID: 31956037 DOI: 10.1016/j.ejca.2019.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prognostic value of tumour-infiltrating lymphocytes (TILs) differs by breast cancer (BC) subtype. The aim of this study was to evaluate TILs in stage III BC in the context of BRCA1/2-like phenotypes and association with outcome and benefit of intensified platinum-based chemotherapy. PATIENTS AND METHODS Patients participated in a randomised controlled trial of adjuvant intensified platinum-based chemotherapy versus conventional anthracycline-based chemotherapy carried out between 1993 and 1999 in stage III BC. Stromal TILs were scored according to International guidelines in these human epidermal growth factor receptor 2 (HER2)-negative tumours. BRCA-profiles were determined using Comparative Genomic Hybridization. RESULTS TIL levels were evaluated in 248 BCs. High TILs were associated with Triple Negative BC (TNBC). BRCA-like tumours harboured higher TILs compared to non-BRCA-like tumours (median TILs of 20% versus 10%, p < 0.01). TIL levels in BRCA1-like tumours were higher compared to BRCA2-like tumours (median TILs of 20% versus 10%, p < 0.001). These correlations remained significant within the oestrogen (ER)-positive subgroup, however not within the TNBC subgroup. In this stage III BC cohort, high TIL level was associated with favourable outcome (TILs per 10% increment, recurrence-free survival (RFS): multivariate hazard ratio (HR) 0.82, 95% confidence interval (CI) 0.71-0.94, p = 0.01; overall survival (OS): multivariate HR 0.80, 95% CI 0.68-0.94, p = 0.01). There was no significant interaction between TILs and benefit of intensified platinum-based chemotherapy. CONCLUSION In this high-risk breast cancer cohort, high TILs were associated with TNBC and BRCA1-like status. Within the ER-positive subgroup, TIL levels were higher in BRCA1-like compared to BRCA2-like tumours. When adjusted for clinical characteristics, TILs were significantly associated with a more favourable outcome in stage III BC patients.
Collapse
MESH Headings
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- BRCA1 Protein/genetics
- BRCA2 Protein/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carboplatin/administration & dosage
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/drug therapy
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/immunology
- Carcinoma, Lobular/pathology
- Chemotherapy, Adjuvant
- Cyclophosphamide/administration & dosage
- Epirubicin/administration & dosage
- Female
- Fluorouracil/administration & dosage
- Follow-Up Studies
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Mutation
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Staging
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Survival Rate
- Thiotepa/administration & dosage
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Leonora de Boo
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Yoni Lubeck
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Antonios Daletzakis
- Biometrics Department, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik Hooijberg
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annelot van Rossum
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Zuzana Loncova
- Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Rieder
- Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Marieke Vollebergh
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marcelo Sobral-Leite
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Koen van de Vijver
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rianne van der Wiel
- Core Facility Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Harm van Tinteren
- Biometrics Department, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pathology, University Medical Centre, Utrecht, the Netherlands
| | - Hugo Mark Horlings
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marleen Kok
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Burotto M, Wilkerson J, Stein WD, Bates SE, Fojo T. Adjuvant and neoadjuvant cancer therapies: A historical review and a rational approach to understand outcomes. Semin Oncol 2019; 46:83-99. [DOI: 10.1053/j.seminoncol.2019.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
|
10
|
Chartron E, Theillet C, Guiu S, Jacot W. Targeting homologous repair deficiency in breast and ovarian cancers: Biological pathways, preclinical and clinical data. Crit Rev Oncol Hematol 2018; 133:58-73. [PMID: 30661659 DOI: 10.1016/j.critrevonc.2018.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/25/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
Mutation or epigenetic silencing of homologous recombination (HR) repair genes is characteristic of a growing proportion of triple-negative breast cancers (TNBCs) and high-grade serous ovarian carcinomas. Defects in HR lead to genome instability, allowing cells to acquire the multiple genetic alterations essential for cancer development. However, this deficiency can also be exploited by using DNA damaging agents or by targeting compensatory repair pathways. A noteworthy example is treatment of TNBC and epithelial ovarian cancer harboring BRCA1/2 germline mutations using platinum salts and/or PARP inhibitors. Dramatic responses to PARP inhibitors may support a wider use in the HR-deficient population beyond those with mutated germline BRCA1 and 2. In this review, we discuss HR deficiency hallmarks as predictive biomarkers for platinum salt and PARP inhibitor sensitivity for selecting patients affected by TNBC or epithelial ovarian cancer who could benefit from these therapeutic options.
Collapse
Affiliation(s)
- Elodie Chartron
- Department of medical oncology, Montpellier Academic Hospital, Montpellier, France
| | - Charles Theillet
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France
| | - Séverine Guiu
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - William Jacot
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France; Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.
| |
Collapse
|
11
|
Kuzhan O, Özet A, Ulutin C, Kömürcü Ş, Arpaci F, Öztürk B, Öztürk M. Survival Benefit with GM-CSF Use after High-Dose Chemotherapy in High-Risk Breast Cancer. TUMORI JOURNAL 2018; 93:550-6. [DOI: 10.1177/030089160709300606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background The role of high-dose chemotherapy in breast cancer has not been fully defined. It has been concluded that new trials should focus on defining potential subgroups that are more likely to benefit from high-dose chemotherapy. We compared survival differences in patients receiving human granulocyte-colony stimulating factor (G-CSF) or granulocyte-monocyte colony stimulating factor (GM-CSF) after high-dose chemotherapy with stem cell support. Methods High-risk non-metastatic breast cancer patients (axillary lymph node involvement more than 8) aged 16 to 65 years and with a performance status ≤1 underwent high-dose chemotherapy with autograft. Written informed consent was obtained from every patient, and the study was approved by the local ethics committee. Results For 54 eligible women, the median follow-up was 41.4 months. The five-year disease-free survival was 45.7%. The five-year projected overall survival rate was 53.9%. Among them, patients who received GM-CSF (n = 12) posttransplant lived longer than the patients who received G-CSF (n = 15) (five year survival rates, 46.6% vs 75%, P <0.050). The patients who received GM-CSF posttransplant had fewer relapses (5 vs 9). However, between the two groups there was no statistically significant difference regarding disease-free survival rates calculated with the Kaplan-Meier method (58.8% vs 40%; P = 0.121). Conclusions Patients receiving GM-CSF posttransplant lived longer and they had fewer relapses than those who received G-CSF. This result merits consideration. The antitumor activity of GM-CSF should be investigated further in prospective randomized trials.
Collapse
Affiliation(s)
- Okan Kuzhan
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| | - Ahmet Özet
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| | - Cüneyt Ulutin
- GATA School of Medicine, Department of Radiation Oncology, Ankara, Etlik, Turkey
| | - Şeref Kömürcü
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| | - Fikret Arpaci
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| | - Bekir Öztürk
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| | - Mustafa Öztürk
- GATA School of Medicine, Department of Medical Oncology, Etlik, Ankara
| |
Collapse
|
12
|
Pajic M, Blatter S, Guyader C, Gonggrijp M, Kersbergen A, Küçükosmanoğlu A, Sol W, Drost R, Jonkers J, Borst P, Rottenberg S. Selected Alkylating Agents Can Overcome Drug Tolerance of G 0-like Tumor Cells and Eradicate BRCA1-Deficient Mammary Tumors in Mice. Clin Cancer Res 2017; 23:7020-7033. [PMID: 28821557 DOI: 10.1158/1078-0432.ccr-17-1279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/08/2017] [Accepted: 08/14/2017] [Indexed: 11/16/2022]
Abstract
Purpose: We aimed to characterize and target drug-tolerant BRCA1-deficient tumor cells that cause residual disease and subsequent tumor relapse.Experimental Design: We studied responses to various mono- and bifunctional alkylating agents in a genetically engineered mouse model for BRCA1/p53-mutant breast cancer. Because of the large intragenic deletion of the Brca1 gene, no restoration of BRCA1 function is possible, and therefore, no BRCA1-dependent acquired resistance occurs. To characterize the cell-cycle stage from which Brca1-/-;p53-/- mammary tumors arise after cisplatin treatment, we introduced the fluorescent ubiquitination-based cell-cycle indicator (FUCCI) construct into the tumor cells.Results: Despite repeated sensitivity to the MTD of platinum drugs, the Brca1-mutated mammary tumors are not eradicated, not even by a frequent dosing schedule. We show that relapse comes from single-nucleated cells delaying entry into the S-phase. Such slowly cycling cells, which are present within the drug-naïve tumors, are enriched in tumor remnants. Using the FUCCI construct, we identified nonfluorescent G0-like cells as the population most tolerant to platinum drugs. Intriguingly, these cells are more sensitive to the DNA-crosslinking agent nimustine, resulting in an increased number of multinucleated cells that lack clonogenicity. This is consistent with our in vivo finding that the nimustine MTD, among several alkylating agents, is the most effective in eradicating Brca1-mutated mouse mammary tumors.Conclusions: Our data show that targeting G0-like cells is crucial for the eradication of BRCA1/p53-deficient tumor cells. This can be achieved with selected alkylating agents such as nimustine. Clin Cancer Res; 23(22); 7020-33. ©2017 AACR.
Collapse
Affiliation(s)
- Marina Pajic
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales, Australia
| | - Sohvi Blatter
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Charlotte Guyader
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maaike Gonggrijp
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ariena Kersbergen
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Aslι Küçükosmanoğlu
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wendy Sol
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rinske Drost
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Piet Borst
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland. .,Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
13
|
Kalimuthu S, Oh JM, Gangadaran P, Zhu L, Lee HW, Jeon YH, Jeong SY, Lee SW, Lee J, Ahn BC. Genetically engineered suicide gene in mesenchymal stem cells using a Tet-On system for anaplastic thyroid cancer. PLoS One 2017; 12:e0181318. [PMID: 28727740 PMCID: PMC5519161 DOI: 10.1371/journal.pone.0181318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 06/29/2017] [Indexed: 12/21/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive malignancy of the thyroid, during which undifferentiated tumors arise from the thyroid follicular epithelium. ATC has a very poor prognosis due to its aggressive behavior and poor response to conventional therapies. Gene-directed enzyme/prodrug therapy using genetically engineered mesenchymal stromal cells (MSC) is a promising therapeutic strategy. The doxycycline (DOX)-controlled Tet inducible system is the most widely utilized regulatory system and could be a useful tool for therapeutic gene-based therapies. For example, use a synthetic "tetracycline-on" switch system to control the expression of the therapeutic gene thymidine kinase, which converts prodrugs to active drugs. The aim of this study was to develop therapeutic MSCs, harboring an inducible suicide gene, and to validate therapeutic gene expression using optical molecular imaging of ATC. We designed the Tet-On system using a retroviral vector expressing herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Mouse bone marrow-derived mesenchymal stromal cells (BM-MSC) were transduced using this system with (MSC-Tet-TK/Fluc2) or without (MSC-TK/Fluc) the Tet-On system. Transduced cells were screened and characterized. Engineered MSCs were co-cultured with ATC (CAL62/Rluc) cells in the presence of the prodrug ganciclovir (GCV) and stimulated with DOX. The efficiency of cell killing monitored by assessing Rluc (CAL62/Rluc) and Fluc (MSC-Tet-TK/Fluc and MSC-TK/Fluc) activities using IVIS imaging. Fluc activity increased in MSC-Tet-TK/Fluc cells in a dose dependent manner following DOX treatment (R2 = 0.95), whereas no signal was observed in untreated cells. eGFP could also be visualized after induction with DOX, and the HSV1-TK protein could be detected by western blotting. In MSC-TK/Fluc cells, the Fluc activity increased with increasing cell number (R2 = 0.98), and eGFP could be visualized by fluorescence microscopy. The Fluc activity and cell viability of MSC-Tet-TK/Fluc and MSC-TK/Fluc cells decreased significantly following GCV treatment. A bystander effect of the therapeutic cells confirmed in co-cultures of CAL62 cells, an anaplastic thyroid cancer cell line, with either MSC-Tet-TK/Fluc cells or MSC-TK/Fluc cells. The Rluc activity in MSC-Tet-TK/Fluc co-cultures, derived from the CAL62/Rluc cells, decreased significantly with GCV treatment of DOX treated cultures, whereas no significant changes were observed in untreated cultures. In addition, the Fluc activity of MSC-Tet-TK/Fluc cells also decreased significantly with DOX treatment whereas no signal was present in untreated cultures. A bystander effect also be demonstrated in co-cultures with MSC-TK/Fluc cells and CAL62/Rluc; both the Rluc activity and the Fluc activity were significantly decreased following GCV treatment. We have successfully developed a Tet-On system of gene-directed enzyme/prodrug delivery using MSCs. We confirmed the therapeutic bystander effect in CAL62/Rluc cells with respect to MSC-Tet-TK/Fluc and MSC-TK/Fluc cells after GCV treatment with and without DOX. Our results confirm the therapeutic efficiency of a suicide gene, with or without the Tet-On system, for ATC therapy. In addition, our findings provide an innovative therapeutic approach for using the Tet-On system to eradicate tumors by simple, repeated administration of MSC-Tet-TK/Fluc cells with DOX and GCV.
Collapse
Affiliation(s)
- Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Liya Zhu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| |
Collapse
|
14
|
Cheng YC, Shi Y, Zhang MJ, Brazauskas R, Hemmer MT, Bishop MR, Nieto Y, Stadtmauer E, Ayash L, Gale RP, Lazarus H, Holmberg L, Lill M, Olsson RF, Wirk BM, Arora M, Hari P, Ueno N. Long-Term Outcome of Inflammatory Breast Cancer Compared to Non-Inflammatory Breast Cancer in the Setting of High-Dose Chemotherapy with Autologous Hematopoietic Cell Transplantation. J Cancer 2017; 8:1009-1017. [PMID: 28529613 PMCID: PMC5436253 DOI: 10.7150/jca.16870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 12/03/2016] [Indexed: 11/21/2022] Open
Abstract
Introduction: Inflammatory breast cancer (IBC) is a rare aggressive form of breast cancer. It is well known that the long-term survival and progression-free survival of IBC are worse than that of non-IBC. We report the long term outcomes of patients with IBC and non-IBC who had undergone high-dose chemotherapy (HDC) with autologous hematopoietic cell transplantation (AHCT). Methods: All 3387 patients with IBC or non-IBC who underwent HDC with AHCT between1990-2002 and registered with CIBMTR were included in this analysis. Transplant-related mortality (TRM), disease relapse/progression, progression-free survival (PFS) and overall survival (OS) were compared between the two cohorts. Multivariate Cox regression model was used to determine the independent impact of stage on outcomes. Results: 527 patients with IBC and 2,860 patients with non-IBC were included; the median age at transplantation (47 vs 46 years old) and median follow-up period in the 2 groups (167 vs 168 months) were similar. The most common conditioning regimen was cyclophosphamide and carboplatin based in both groups (54% in IBC and 50% in non-IBC). AHCT was well tolerated in both groups. TRM was similar in both groups (one year TRM was 2% for IBC and 3% for non-IBC, p=0.16). The most common cause of death was disease progression or relapse (81% in IBC and 75% in non-IBC). The median survival for both IBC and non-IBC was the same at 40 months. The PFS at 10 years was 27% (95% CI: 23-31%) for IBC and 24% (95% CI: 22-26%) for non-IBC (p=0.21), and the OS at 10 years was 31% (95% CI: 27-35%) for IBC and 28% (95% CI: 26-30%) for non-IBC (p=0.16). In univariate analysis, patients with stage III IBC and no active diseases at transplantation had lower PFS and OS than that in non-IBC. In multivariate analysis, controlling for age, disease status at AHCT, hormonal receptor status, time from diagnosis to AHCT, and performance status at AHCT, patients with stage III IBC had higher mortality (HR 1.16, 95% CI: 1-1.34, p= 0.0459), worse PFS (HR: 1.17, 95% CI: 1.01-1.36, p= 0.0339) and higher risk of disease relapse/progression (HR: 1.24, 95% CI: 1.06-1.45, p= 0.0082) as compared to stage III non-IBC. Amongst all patients a higher stage disease was associated with worse PFS, OS and disease relapse/progression. Conclusions: Long-term outcomes of stage III IBC patients who underwent AHCT were poorer than that in non-IBC patients confirming that the poor prognosis of IBC even in the setting of HDC with AHCT.
Collapse
Affiliation(s)
| | - Yushu Shi
- Medical College of Wisconsin, Milwaukee, WI
| | - Mei-Jie Zhang
- CIBMTR(Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.,Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI
| | - Ruta Brazauskas
- CIBMTR(Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael T Hemmer
- CIBMTR(Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Edward Stadtmauer
- Abramson Cancer Center University of Pennsylvania Medical Center, Philadelphia, PA
| | - Lois Ayash
- Karmanos Cancer Institute, Detroit, MI; Division of Hematology, Oncology, Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Hillard Lazarus
- Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, OH
| | | | | | - Richard F Olsson
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Baldeep Mona Wirk
- Division of Bone Marrow Transplant, Seattle Cancer Care Alliance, Seattle, WA
| | - Mukta Arora
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN
| | - Parameswaran Hari
- CIBMTR(Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Naoto Ueno
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
15
|
Van Pham P. Stem cell drugs: the next generation of pharmaceutical products. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0047-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
16
|
van Harten WH, Retèl VP. Innovations that reach the patient: early health technology assessment and improving the chances of coverage and implementation. Ecancermedicalscience 2016; 10:683. [PMID: 27899956 PMCID: PMC5102688 DOI: 10.3332/ecancer.2016.683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Indexed: 11/06/2022] Open
Abstract
With growing concerns for the sustainability of the financial burden that health care-and especially cancer services-poses on the national budgets, the role of health economic analyses in coverage decisions is likely to grow. One of the strategies for the biomedical research field-also in oncology research-to foster coverage and health system implementation, is to anticipate this new role and to involve health technology assessment techniques earlier in various stages of translational research. In this article, we elaborate on the early involvement of health technology assessment in translational research and the concept of Coverage with Evidence Development in The Netherlands Cancer Institute and give two case examples that are currently ongoing: (1) tumour infiltrating lymphocytes therapy for metastatic melanoma; and (2) high-dose chemotherapy for BRCA1-like subgroup in triple-negative breast cancer. We conclude with recommendations for institutional policy.
Collapse
Affiliation(s)
- W H van Harten
- The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; University of Twente, Department of Healthcare Technology and Services Research, PO Box 217, 7500 AE Enschede, The Netherlands; Rijnstate Hospital, PO Box 9555, 6800 TA Arnhem, The Netherlands
| | - V P Retèl
- The Netherlands Cancer Institute, PO Box 90203, 1006 BE Amsterdam, The Netherlands; University of Twente, Department of Healthcare Technology and Services Research, PO Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
17
|
Salek R, Bayatmokhtari N, Homaei Shandiz F, ShahidSales S. The Results of Chemotherapy with Two Variants of Intravenous CMF in Patients with Early Stage Breast Carcinoma; Does Dose Density Matter? Breast J 2016; 22:623-629. [PMID: 27540897 DOI: 10.1111/tbj.12652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
No direct comparisons can be made in early stages of breast cancer, between the intravenous combinations of: cyclophosphamide, methotrexate, and fluorouracil; named modified versions of CMF with the classical oral version of CMF. Since these modifications have different dose intensities and densities, the outcomes for their subsequent treatments may be varied, and not produce the same results. Despite that, classical CMF has been commonly replaced with intravenous modifications. This study aimed to assess the results of treatment with two common intravenous modification of CMF chemotherapy; to represent the most effective and successful substitute of classical CMF. Five hundred patients in two groups were eligible to take part in the experiment. For two hundred and twenty-nine patients in the group CMF 1&8, chemotherapy was administered intravenously on days 1 and 8 every 28 days for six cycles consisting of: cyclophosphamide 600 mg/m2 , methotrexate 40 mg/m2 , fluorouracil 600 mg/m2 . In the group CMF 1 which consisted of 271 patients, chemotherapy was administered with all the same drugs and doses, however, it was only administered on day 1 and repeated at 21-day intervals for six cycles. Overall survival (OS), disease-free survival (DFS), the prognostic factors and other probable interventional factors were then compared between the two groups. The 5-year OS rate of 87.5% and 10-year OS rate of 82% in the group CMF 1&8 were statistically significantly better than 5-year OS of 84% and 10-year OS of 61.5% in the group CMF 1 (p = 0.01). The 5-year and 10-year DFS rates were 76% and 60% respectively, in the group CMF 1&8 compared with 77% and 54% respectively in the group CMF 1 (p = 0.8). Two groups were comparable regarding their distribution of different prognostic factors and other probable interventional factors. Considering 30% higher dose density of drugs in the protocol of CMF 1&8, the improving outcome can be related to the efficacy of dose-dense chemotherapy. Therefore, this intravenous modification is the better substitute of classical CMF.
Collapse
Affiliation(s)
- Roham Salek
- Clinical Radiation Oncology, Faculty of Medicine, Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Bayatmokhtari
- Clinical Radiation Oncology, Sabzevar University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Homaei Shandiz
- Clinical Radiation Oncology, Faculty of Medicine, Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Clinical Radiation Oncology, Faculty of Medicine, Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Miquel-Cases A, Retèl VP, van Harten WH, Steuten LMG. Decisions on Further Research for Predictive Biomarkers of High-Dose Alkylating Chemotherapy in Triple-Negative Breast Cancer: A Value of Information Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2016; 19:419-430. [PMID: 27325334 DOI: 10.1016/j.jval.2016.01.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 01/28/2016] [Accepted: 01/31/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVES To inform decisions about the design and priority of further studies of emerging predictive biomarkers of high-dose alkylating chemotherapy (HDAC) in triple-negative breast cancer (TNBC) using value-of-information analysis. METHODS A state transition model compared treating women with TNBC with current clinical practice and four biomarker strategies to personalize HDAC: 1) BRCA1-like profile by array comparative genomic hybridization (aCGH) testing; 2) BRCA1-like profile by multiplex ligation-dependent probe amplification (MLPA) testing; 3) strategy 1 followed by X-inactive specific transcript gene (XIST) and tumor suppressor p53 binding protein (53BP1) testing; and 4) strategy 2 followed by XIST and 53BP1 testing, from a Dutch societal perspective and a 20-year time horizon. Input data came from literature and expert opinions. We assessed the expected value of partial perfect information, the expected value of sample information, and the expected net benefit of sampling for potential ancillary studies of an ongoing randomized controlled trial (RCT; NCT01057069). RESULTS The expected value of partial perfect information indicated that further research should be prioritized to the parameter group including "biomarkers' prevalence, positive predictive value (PPV), and treatment response rates (TRRs) in biomarker-negative patients and patients with TNBC" (€639 million), followed by utilities (€48 million), costs (€40 million), and transition probabilities (TPs) (€30 million). By setting up four ancillary studies to the ongoing RCT, data on 1) TP and MLPA prevalence, PPV, and TRR; 2) aCGH and aCGH/MLPA plus XIST and 53BP1 prevalence, PPV, and TRR; 3) utilities; and 4) costs could be simultaneously collected (optimal size = 3000). CONCLUSIONS Further research on predictive biomarkers for HDAC should focus on gathering data on TPs, prevalence, PPV, TRRs, utilities, and costs from the four ancillary studies to the ongoing RCT.
Collapse
Affiliation(s)
- Anna Miquel-Cases
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands.
| | - Lotte M G Steuten
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
19
|
Farquhar C, Marjoribanks J, Lethaby A, Azhar M. High-dose chemotherapy and autologous bone marrow or stem cell transplantation versus conventional chemotherapy for women with early poor prognosis breast cancer. Cochrane Database Syst Rev 2016; 2016:CD003139. [PMID: 27200512 PMCID: PMC8078206 DOI: 10.1002/14651858.cd003139.pub3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Overall survival rates are disappointing for women with early poor prognosis breast cancer. Autologous transplantation of bone marrow or peripheral stem cells (in which the woman is both donor and recipient) has been considered a promising technique because it permits use of much higher doses of chemotherapy. OBJECTIVES To compare the effectiveness and safety of high-dose chemotherapy and autograft (either autologous bone marrow or stem cell transplantation) with conventional chemotherapy for women with early poor prognosis breast cancer. SEARCH METHODS We searched the Cochrane Breast Cancer Group Specialised Register, MEDLINE (1966 to October 2015), EMBASE (1980 to October 2015), the World Health Organization's International Clinical Trials Registry Search Platform, and ClinicalTrials.gov on the 21 October 2015. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing high-dose chemotherapy and autograft (bone marrow transplant or stem cell rescue) versus chemotherapy without autograft for women with early poor prognosis breast cancer. DATA COLLECTION AND ANALYSIS Two review authors selected RCTs, independently extracted data and assessed risks of bias. We combined data using a Mantel-Haenszel fixed-effect model to calculate pooled risk ratios (RRs) and 95% confidence intervals (CIs). We assessed the quality of the evidence using GRADE methods. Outcomes were survival rates, toxicity and quality of life. MAIN RESULTS We included 14 RCTs of 5600 women randomised to receive high-dose chemotherapy and autograft (bone marrow transplant or stem cell rescue) versus chemotherapy without autograft for women with early poor prognosis breast cancer. The studies were at low risk of bias in most areas.There is high-quality evidence that high-dose chemotherapy does not increase the likelihood of overall survival at any stage of follow-up (at three years: RR 1.02, 95% CI 0.95 to 1.10, 3 RCTs, 795 women, I² = 56%; at five years: RR 1.00, 95% CI 0.96 to 1.04, 9 RCTs, 3948 women, I² = 0%; at six years: RR 0.94, 95% CI 0.81 to 1.08, 1 RCT, 511 women; at eight years: RR1.17, 95% CI 0.95 to 1.43, 1 RCT, 344 women; at 12 years: RR 1.18, 95% CI 0.99 to 1.42, 1 RCT, 382 women).There is high-quality evidence that high-dose chemotherapy improves the likelihood of event-free survival at three years (RR 1.19, 95% CI 1.06 to 1.34, 3 RCTs, 795 women, I² = 56%) but this effect was no longer apparent at longer duration of follow-up (at five years: RR 1.04, 95% CI 0.99 to 1.09, 9 RCTs, 3948 women, I² = 14%; at six years RR 1.04, 95% CI 0.87 to 1.24, 1 RCT, 511 women; at eight years: RR 1.27, 95% CI 0.99 to 1.64, 1 RCT, 344 women; at 12 years: RR 1.18, 95% CI 0.95 to 1.45, 1 RCT, 382 women).Treatment-related deaths were much more frequent in the high-dose arm (RR 7.97, 95% CI 3.99 to 15.92, 14 RCTs, 5600 women, I² = 12%, high-quality evidence) and non-fatal morbidity was also more common and more severe in the high-dose group. There was little or no difference between the groups in the incidence of second cancers at four to nine years' median follow-up (RR 1.25, 95% CI 0.90 to 1.73, 7 RCTs, 3423 women, I² = 0%, high-quality evidence). Women in the high-dose group reported significantly worse quality-of-life scores immediately after treatment, but there were few statistically significant differences between the groups by one year.The primary studies were at low risk of bias in most areas, and the evidence was assessed using GRADE methods and rated as high quality for all comparisons. AUTHORS' CONCLUSIONS There is high-quality evidence of increased treatment-related mortality and little or no increase in survival by using high-dose chemotherapy with autograft for women with early poor prognosis breast cancer.
Collapse
Affiliation(s)
- Cindy Farquhar
- University of AucklandDepartment of Obstetrics and GynaecologyFMHS Park RoadGraftonAucklandNew Zealand1003
| | - Jane Marjoribanks
- University of AucklandDepartment of Obstetrics and GynaecologyFMHS Park RoadGraftonAucklandNew Zealand1003
| | - Anne Lethaby
- University of AucklandDepartment of Obstetrics and GynaecologyFMHS Park RoadGraftonAucklandNew Zealand1003
| | - Maimoona Azhar
- Royal College of Surgeons in IrelandDepartment of Surgery123 St. Stephen's GreenDublin 2Ireland
| | | |
Collapse
|
20
|
A Uridine Glucuronosyltransferase 2B7 Polymorphism Predicts Epirubicin Clearance and Outcomes in Early-Stage Breast Cancer. Clin Breast Cancer 2016; 16:139-44.e1-3. [DOI: 10.1016/j.clbc.2015.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/31/2015] [Accepted: 09/11/2015] [Indexed: 11/22/2022]
|
21
|
Stouten-Kemperman MM, de Ruiter MB, Koppelmans V, Boogerd W, Reneman L, Schagen SB. Neurotoxicity in breast cancer survivors ≥10 years post-treatment is dependent on treatment type. Brain Imaging Behav 2016; 9:275-84. [PMID: 24858488 DOI: 10.1007/s11682-014-9305-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Adjuvant chemotherapy (CT) for breast cancer (BC) is associated with very late side-effects on brain function and structure. However, little is known about neurotoxicity of specific treatment regimens. To compare neurotoxicity profiles after different treatment strategies, we used neurocognitive testing and multimodality MRI in BC survivors randomized to high-dose (HI), conventional-dose (CON-) CT or radiotherapy (RT) only and a healthy control (HC) group. BC survivors who received CON-CT (n = 20) and HC (n = 20) were assessed using a neurocognitive test battery and multimodality MRI including 3D-T1, Diffusion Tensor Imaging (DTI) and 1H-MR spectroscopy (1H-MRS) to measure various aspects of cerebral white (WM) and gray matter (GM). Data were compared to previously assessed groups of BC survivors who received HI-CT (n = 17) and RT-only (n = 15). Testing took place on average 11.5 years post-CT. 3D-T1 showed focal GM volume reductions both for HI-CT and CON-CT compared to RT-only (p < .004). DTI-derived mean diffusivity and 1H-MRS derived N-acetyl aspartate showed WM injury specific to HI-CT but not CON-CT (p < .05). Residual effects were revealed in the RT-only group compared to HC on MRI and neurocognitive measurements (p < .05). Ten years after adjuvant CT for BC lower cerebral GM volume was found in HI as well as CON-CT BC survivors whereas injury to WM is restricted to HI-CT. This might indicate that WM brain changes after BC treatment may show more pronounced (partial) recovery than GM. Furthermore, our results suggest residual neurotoxicity in the RT-only group, which warrants further investigation.
Collapse
Affiliation(s)
- Myrle M Stouten-Kemperman
- Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
22
|
Zhou M, Zhao J, Tian M, Song S, Zhang R, Gupta S, Tan D, Shen H, Ferrari M, Li C. Radio-photothermal therapy mediated by a single compartment nanoplatform depletes tumor initiating cells and reduces lung metastasis in the orthotopic 4T1 breast tumor model. NANOSCALE 2015; 7:19438-47. [PMID: 26376843 PMCID: PMC4993020 DOI: 10.1039/c5nr04587h] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Tumor Initiating Cells (TICs) are resistant to radiotherapy and chemotherapy, and are believed to be responsible for tumor recurrence and metastasis. Combination therapies can overcome the limitation of conventional cancer treatments, and have demonstrated promising application in the clinic. Here, we show that dual modality radiotherapy (RT) and photothermal therapy (PTT) mediated by a single compartment nanosystem copper-64-labeled copper sulfide nanoparticles ([(64)Cu]CuS NPs) could suppress breast tumor metastasis through eradication of TICs. Positron electron tomography (PET) imaging and biodistribution studies showed that more than 90% of [(64)Cu]CuS NPs was retained in subcutaneously grown BT474 breast tumor 24 h after intratumoral (i.t.) injection, indicating the NPs are suitable for the combination therapy. Combined RT/PTT therapy resulted in significant tumor growth delay in the subcutaneous BT474 breast cancer model. Moreover, RT/PTT treatment significantly prolonged the survival of mice bearing orthotopic 4T1 breast tumors compared to no treatment, RT alone, or PTT alone. The RT/PTT combination therapy significantly reduced the number of tumor nodules in the lung and the formation of tumor mammospheres from treated 4T1 tumors. No obvious side effects of the CuS NPs were noted in the treated mice in a pilot toxicity study. Taken together, our data support the feasibility of a therapeutic approach for the suppression of tumor metastasis through localized RT/PTT therapy.
Collapse
Affiliation(s)
- Min Zhou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA. and The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Zhao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Mei Tian
- The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaoli Song
- Department of Nuclear Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Rui Zhang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Sanjay Gupta
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Dongfeng Tan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, The Methodist Hospital System Research Institute, Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, The Methodist Hospital System Research Institute, Houston, TX 77030, USA
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| |
Collapse
|
23
|
Schouten PC, Vollebergh MA, Opdam M, Jonkers M, Loden M, Wesseling J, Hauptmann M, Linn SC. High XIST and Low 53BP1 Expression Predict Poor Outcome after High-Dose Alkylating Chemotherapy in Patients with a BRCA1-like Breast Cancer. Mol Cancer Ther 2015; 15:190-8. [DOI: 10.1158/1535-7163.mct-15-0470] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/06/2015] [Indexed: 11/16/2022]
|
24
|
Biesma HD, Schouten PC, Lacle MM, Sanders J, Brugman W, Kerkhoven R, Mandjes I, van der Groep P, van Diest PJ, Linn SC. Copy number profiling by array comparative genomic hybridization identifies frequently occurring BRCA2-like male breast cancer. Genes Chromosomes Cancer 2015; 54:734-44. [DOI: 10.1002/gcc.22284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/25/2015] [Indexed: 11/08/2022] Open
Affiliation(s)
- Hedde D. Biesma
- Department of Molecular Pathology; Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Philip C. Schouten
- Department of Molecular Pathology; Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Miangela M. Lacle
- Department of Pathology; University Medical Center Utrecht; The Netherlands
| | - Joyce Sanders
- Department of Pathology; Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Wim Brugman
- Genomics Core Facility, Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Ron Kerkhoven
- Genomics Core Facility, Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Ingrid Mandjes
- Data Center, Netherlands Cancer Institute; Amsterdam The Netherlands
| | | | - Paul J. van Diest
- Department of Pathology; University Medical Center Utrecht; The Netherlands
| | - Sabine C. Linn
- Department of Molecular Pathology; Netherlands Cancer Institute; Amsterdam The Netherlands
- Department of Pathology; University Medical Center Utrecht; The Netherlands
- Department of Medical Oncology; Netherlands Cancer Institute; Amsterdam The Netherlands
| |
Collapse
|
25
|
Schouten PC, Grigoriadis A, Kuilman T, Mirza H, Watkins JA, Cooke SA, van Dyk E, Severson TM, Rueda OM, Hoogstraat M, Verhagen CVM, Natrajan R, Chin SF, Lips EH, Kruizinga J, Velds A, Nieuwland M, Kerkhoven RM, Krijgsman O, Vens C, Peeper D, Nederlof PM, Caldas C, Tutt AN, Wessels LF, Linn SC. Robust BRCA1-like classification of copy number profiles of samples repeated across different datasets and platforms. Mol Oncol 2015; 9:1274-86. [PMID: 25825120 PMCID: PMC5528812 DOI: 10.1016/j.molonc.2015.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/01/2015] [Accepted: 03/11/2015] [Indexed: 11/30/2022] Open
Abstract
Breast cancers with BRCA1 germline mutation have a characteristic DNA copy number (CN) pattern. We developed a test that assigns CN profiles to be 'BRCA1-like' or 'non-BRCA1-like', which refers to resembling a BRCA1-mutated tumor or resembling a tumor without a BRCA1 mutation, respectively. Approximately one third of the BRCA1-like breast cancers have a BRCA1 mutation, one third has hypermethylation of the BRCA1 promoter and one third has an unknown reason for being BRCA1-like. This classification is indicative of patients' response to high dose alkylating and platinum containing chemotherapy regimens, which targets the inability of BRCA1 deficient cells to repair DNA double strand breaks. We investigated whether this classification can be reliably obtained with next generation sequencing and copy number platforms other than the bacterial artificial chromosome (BAC) array Comparative Genomic Hybridization (aCGH) on which it was originally developed. We investigated samples from 230 breast cancer patients for which a CN profile had been generated on two to five platforms, comprising low coverage CN sequencing, CN extraction from targeted sequencing panels (CopywriteR), Affymetrix SNP6.0, 135K/720K oligonucleotide aCGH, Affymetrix Oncoscan FFPE (MIP) technology, 3K BAC and 32K BAC aCGH. Pairwise comparison of genomic position-mapped profiles from the original aCGH platform and other platforms revealed concordance. For most cases, biological differences between samples exceeded the differences between platforms within one sample. We observed the same classification across different platforms in over 80% of the patients and kappa values of at least 0.36. Differential classification could be attributed to CN profiles that were not strongly associated to one class. In conclusion, we have shown that the genomic regions that define our BRCA1-like classifier are robustly measured by different CN profiling technologies, providing the possibility to retro- and prospectively investigate BRCA1-like classification across a wide range of CN platforms.
Collapse
Affiliation(s)
- Philip C Schouten
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anita Grigoriadis
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom
| | - Thomas Kuilman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hasan Mirza
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom
| | - Johnathan A Watkins
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom
| | - Saskia A Cooke
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom
| | - Ewald van Dyk
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tesa M Severson
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Oscar M Rueda
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Marlous Hoogstraat
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands; Netherlands Center for Personalized Cancer Treatment, Utrecht, The Netherlands
| | - Caroline V M Verhagen
- Division of Biological Stress Response, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Esther H Lips
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Janneke Kruizinga
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Arno Velds
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marja Nieuwland
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ron M Kerkhoven
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Oscar Krijgsman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Conchita Vens
- Division of Biological Stress Response, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniel Peeper
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petra M Nederlof
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK; Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical, Research Centre, Cambridge University Hospitals NHS, Cambridge, UK
| | - Andrew N Tutt
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom
| | - Lodewyk F Wessels
- Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands; Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, The Netherlands
| | - Sabine C Linn
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands; Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Sánchez-Aguilera A, Arranz L, Martín-Pérez D, García-García A, Stavropoulou V, Kubovcakova L, Isern J, Martín-Salamanca S, Langa X, Skoda RC, Schwaller J, Méndez-Ferrer S. Estrogen signaling selectively induces apoptosis of hematopoietic progenitors and myeloid neoplasms without harming steady-state hematopoiesis. Cell Stem Cell 2015; 15:791-804. [PMID: 25479752 DOI: 10.1016/j.stem.2014.11.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/08/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
Estrogens are potent regulators of mature hematopoietic cells; however, their effects on primitive and malignant hematopoietic cells remain unclear. Using genetic and pharmacological approaches, we observed differential expression and function of estrogen receptors (ERs) in hematopoietic stem cell (HSC) and progenitor subsets. ERα activation with the selective ER modulator (SERM) tamoxifen induced apoptosis in short-term HSCs and multipotent progenitors. In contrast, tamoxifen induced proliferation of quiescent long-term HSCs, altered the expression of self-renewal genes, and compromised hematopoietic reconstitution after myelotoxic stress, which was reversible. In mice, tamoxifen treatment blocked development of JAK2(V617F)-induced myeloproliferative neoplasm in vivo, induced apoptosis of human JAK2(V617F+) HSPCs in a xenograft model, and sensitized MLL-AF9(+) leukemias to chemotherapy. Apoptosis was selectively observed in mutant cells, and tamoxifen treatment only had a minor impact on steady-state hematopoiesis in disease-free animals. Together, these results uncover specific regulation of hematopoietic progenitors by estrogens and potential antileukemic properties of SERMs.
Collapse
Affiliation(s)
- Abel Sánchez-Aguilera
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.
| | - Lorena Arranz
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Daniel Martín-Pérez
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Andrés García-García
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Vaia Stavropoulou
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Lucia Kubovcakova
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Joan Isern
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Sandra Martín-Salamanca
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Xavier Langa
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Radek C Skoda
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jürg Schwaller
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Simón Méndez-Ferrer
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.
| |
Collapse
|
27
|
Early stage cost-effectiveness analysis of a BRCA1-like test to detect triple negative breast cancers responsive to high dose alkylating chemotherapy. Breast 2015; 24:397-405. [PMID: 25937263 DOI: 10.1016/j.breast.2015.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Triple negative breast cancers (TNBC) with a BRCA1-like profile may benefit from high dose alkylating chemotherapy (HDAC). This study examines whether BRCA1-like testing to target effective HDAC in TNBC patients can be more cost-effective than treating all patients with standard chemotherapy. Additionally, we estimated the minimum required prevalence of BRCA1-like and the required positive predictive value (PPV) for a BRCA1-like test to become cost-effective. METHODS Our Markov model compared 1) the incremental costs; 2) the incremental number of respondents; 3) the incremental number of Quality Adjusted Life Years (QALYs); and 4) the incremental cost-effectiveness ratio (ICER) of treating TNBC women with personalized HDAC based on BRCA1-like testing vs. standard chemotherapy, from a Dutch societal perspective and a 20-year time horizon, using probabilistic sensitivity analysis. Furthermore, we performed one-way sensitivity analysis (SA) to all model parameters, and two-way SA to prevalence and PPV. Data were obtained from a current trial (NCT01057069), published literature and expert opinions. RESULTS BRCA1-like testing to target effective HDAC would presently not be cost-effective at a willingness-to-pay threshold of €80.000/QALY (€81.981/QALY). SAs show that PPV drives the ICER changes. Lower bounds for the prevalence and the PPV were found to be 58.5% and 73.0% respectively. CONCLUSION BRCA1-like testing to target effective HDAC treatment in TNBC patients is currently not cost-effective at a willingness-to-pay of €80.000/QALY, but it can be when a minimum PPV of 73% is obtained in clinical practice. This information can help test developers and clinicians in decisions on further research and development of BRCA1-like tests.
Collapse
|
28
|
Vaxman I, Ram R, Gafter-Gvili A, Vidal L, Yeshurun M, Lahav M, Shpilberg O. Secondary malignancies following high dose therapy and autologous hematopoietic cell transplantation-systematic review and meta-analysis. Bone Marrow Transplant 2015; 50:706-14. [PMID: 25665042 DOI: 10.1038/bmt.2014.325] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 11/09/2022]
Abstract
We performed a systematic review and meta-analysis of randomized controlled trials comparing autologous hematopoietic cell transplantation (HCT) with other treatment modalities to analyze the risk for various secondary malignancies (SMs). Relative risks (RR) with 95% confidence intervals were estimated and pooled. Our search yielded 36 trials. The median follow-up was 55 (range 12-144) months. Overall, the RR for developing SMs was 1.23 ((0.97-1.55), I(2)=4%, 9870 patients). Subgroup analysis of trials assessing TBI-containing preparative regimens and of patients with baseline lymphoproliferative diseases, showed there was a higher risk for SMs in patients given autografts (RR=1.61 (1.05-2.48), I(2)=14%, 2218 patients and RR=1.62 (1.12-2.33), I(2)=22%, 3343 patients, respectively). Among all patients, there was a higher rate of myelodysplastic syndrome MDS/AML in patients given HCT compared with other treatments (RR=1.71 (1.18-2.48), I(2)=0%, 8778 patients). The risk of secondary solid malignancies was comparable in the short term between patients given HCT and patients given other treatments (RR=0.95 (0.67-1.32), I(2)=0%, 5925 patients). We conclude that overall the risk of secondary MDS/AML is higher in patients given autologous HCT compared with other treatments. In the subgroup of patients given a TBI-based regimen and in those with a baseline lymphoproliferative disease, there was a higher risk of overall SMs.
Collapse
Affiliation(s)
- I Vaxman
- 1] Medicine A, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel [2] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - R Ram
- 1] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel [2] BMT Unit, Sourasky Medical Center, Tel Aviv, Israel
| | - A Gafter-Gvili
- 1] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel [2] Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Tel Aviv, Israel
| | - L Vidal
- 1] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel [2] Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Tel Aviv, Israel
| | - M Yeshurun
- 1] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel [2] Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Tel Aviv, Israel
| | - M Lahav
- 1] Medicine A, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel [2] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - O Shpilberg
- 1] Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel [2] Institute of Hematology, Assuta Medical Center, Tel Aviv, Israel
| |
Collapse
|
29
|
Abstract
Although adjuvant chemotherapy (CT) for breast cancer (BC) is associated with very late side-effects on cognition and brain function, studies on adverse effects of specific treatment regimens are scarce. Here, neurotoxicity profiles after different treatment strategies were compared in BC survivors randomized to high-dose (HI) or conventional-dose (CON-) CT, in women treated with radiotherapy (RT) -only and a healthy control (HC) group. We administered a neurocognitive test battery, a planning fMRI task (Tower of London) and episodic memory fMRI task (Paired Associates paradigm) in BC survivors who received CON-CT (n=24) and HC (n=27). Data were compared to BC survivors who received HI-CT (n=17) and RT-only (n=15) and who were previously assessed. Testing took place ±11.5 years post-CT. Furthermore, neurocognitive data were compared to neurocognitive data acquired ≤2 years post-treatment. Cognitive assessment revealed sustained cognitive decline in 10.5% of HI-CT, 8.3% of CON-CT, 6.7% of RT-only patients and 0% in the HC. Hypoactivation was found in task-related prefrontal and parietal areas for both CT-groups versus RT-only, with HI-CT showing more pronounced hypoactivation than CON-CT, combined with worse task performance. RT-only survivors performed at a similar level to HC while showing hyperactivation in task-related brain areas. Long after treatment, CT is associated with cognitive problems and task-related hypoactivation that depend on the specific cytotoxic regimen. This worse performance in patients who received CT could be explained by impaired brain functioning that is more severe with more intense CT.
Collapse
|
30
|
Schouten PC, Marmé F, Aulmann S, Sinn HP, van Essen HF, Ylstra B, Hauptmann M, Schneeweiss A, Linn SC. Breast cancers with a BRCA1-like DNA copy number profile recur less often than expected after high-dose alkylating chemotherapy. Clin Cancer Res 2014; 21:763-70. [PMID: 25480832 DOI: 10.1158/1078-0432.ccr-14-1894] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Breast cancers in carriers of inactivating mutations of the BRCA1 gene carry a specific DNA copy-number signature ("BRCA1-like"). This signature is shared with cancers that inactivate BRCA1 through other mechanisms. Because BRCA1 is important in repair of DNA double-strand breaks through error-free homologous recombination, patients with a BRCA1-like tumor may benefit from high-dose alkylating (HD) chemotherapy, which induces DNA double-strand breaks. EXPERIMENTAL DESIGN We investigated a single institution cohort of high-risk patients that received tandem HD chemotherapy schedule comprising ifosfamide, epirubicin, and carboplatin or conventional chemotherapy. We classified copy-number profiles to be BRCA1-like or non-BRCA1-like and analyzed clinical associations and performed survival analysis with a treatment by biomarker interaction design. RESULTS BRCA1-like status associated with high-grade and triple-negative breast cancers. BRCA1-like cases benefitted from the HD compared with a conventional regimen on disease-free survival (DFS): [hazard ratio (HR), 0.05; 95% confidence interval (CI), 0.01-0.38; P = 0.003]; distant DFS (DDFS): (HR, 0.06; 95% CI, 0.01-0.43; P = 0.01); and overall survival (OS; HR, 0.15; 95% CI, 0.03-0.83; P = 0.03) after correction for prognostic factors. No such benefit was observed in the non-BRCA1-like cases on DFS (HR, 0.74; 95% CI, 0.38-1.46; P = 0.39), DDFS (HR, 0.79; 95% CI, 0.41-1.52; P = 0.47), and OS (HR, 0.93; 95% CI, 0.52-1.64; P = 0.79). The P values for interaction were 0.01 (DFS), 0.01 (DDFS), and 0.045 (OS). CONCLUSIONS BRCA1-like tumors recurred significantly less often after HD than conventional chemotherapy. BRCA1-like copy-number profile classification may be a predictive marker for HD alkylating chemotherapy.
Collapse
Affiliation(s)
- Philip C Schouten
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frederik Marmé
- Universitäts-Frauenklinik, University of Heidelberg, Heidelberg, Germany. National Center for Tumour Diseases, Heidelberg, Germany
| | - Sebastian Aulmann
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Hendrik F van Essen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Michael Hauptmann
- Department of Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andreas Schneeweiss
- Universitäts-Frauenklinik, University of Heidelberg, Heidelberg, Germany. National Center for Tumour Diseases, Heidelberg, Germany
| | - Sabine C Linn
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
31
|
Labots M, Schütte LM, van der Mijn JC, Pham TV, Jiménez CR, Verheul HMW. Mass spectrometry-based serum and plasma peptidome profiling for prediction of treatment outcome in patients with solid malignancies. Oncologist 2014; 19:1028-39. [PMID: 25187478 DOI: 10.1634/theoncologist.2014-0101] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Treatment selection tools are needed to enhance the efficacy of targeted treatment in patients with solid malignancies. Providing a readout of aberrant signaling pathways and proteolytic events, mass spectrometry-based (MS-based) peptidomics enables identification of predictive biomarkers, whereas the serum or plasma peptidome may provide easily accessible signatures associated with response to treatment. In this systematic review, we evaluate MS-based peptide profiling in blood for prompt clinical implementation. METHODS PubMed and Embase were searched for studies using a syntax based on the following hierarchy: (a) blood-based matrix-assisted or surface-enhanced laser desorption/ionization time-of-flight MS peptide profiling (b) in patients with solid malignancies (c) prior to initiation of any treatment modality, (d) with availability of outcome data. RESULTS Thirty-eight studies were eligible for review; the majority were performed in patients with non-small cell lung cancer (NSCLC). Median classification prediction accuracy was 80% (range: 66%-93%) in 11 models from 14 studies reporting an MS-based classification model. A pooled analysis of 9 NSCLC studies revealed clinically significant median progression-free survival in patients classified as "poor outcome" and "good outcome" of 2.0 ± 1.06 months and 4.6 ± 1.60 months, respectively; median overall survival was also clinically significant at 4.01 ± 1.60 months and 10.52 ± 3.49 months, respectively. CONCLUSION Pretreatment MS-based serum and plasma peptidomics have shown promising results for prediction of treatment outcome in patients with solid tumors. Limited sample sizes and absence of signature validation in many studies have prohibited clinical implementation thus far. Our pooled analysis and recent results from the PROSE study indicate that this profiling approach enables treatment selection, but additional prospective studies are warranted.
Collapse
Affiliation(s)
- Mariette Labots
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Lisette M Schütte
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Thang V Pham
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Connie R Jiménez
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Vollebergh MA, Klijn C, Schouten PC, Wesseling J, Israeli D, Ylstra B, Wessels LF, Jonkers J, Linn SC. Lack of genomic heterogeneity at high-resolution aCGH between primary breast cancers and their paired lymph node metastases. PLoS One 2014; 9:e103177. [PMID: 25083859 PMCID: PMC4118860 DOI: 10.1371/journal.pone.0103177] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 06/29/2014] [Indexed: 11/18/2022] Open
Abstract
Lymph-node metastasis (LNM) predict high recurrence rates in breast cancer patients. Systemic treatment aims to eliminate (micro)metastatic cells. However decisions regarding systemic treatment depend largely on clinical and molecular characteristics of primary tumours. It remains, however, unclear to what extent metastases resemble the cognate primary breast tumours, especially on a genomic level, and as such will be eradicated by the systemic therapy chosen. In this study we used high-resolution aCGH to investigate DNA copy number differences between primary breast cancers and their paired LNMs. To date, no recurrent LNM-specific genomic aberrations have been identified using array comparative genomic hybridization (aCGH) analysis. In our study we employ a high-resolution platform and we stratify on different breast cancer subtypes, both aspects that might have underpowered previously performed studies.To test the possibility that genomic instability in triple-negative breast cancers (TNBCs) might cause increased random and potentially also recurrent copy number aberrations (CNAs) in their LNMs, we studied 10 primary TNBC–LNM pairs and 10 ER-positive (ER+) pairs and verified our findings adding additionally 5 TNBC-LNM and 22 ER+-LNM pairs. We found that all LNMs clustered nearest to their matched tumour except for two cases, of which one was due to the presence of two distinct histological components in one tumour. We found no significantly altered CNAs between tumour and their LNMs in the entire group or in the subgroups. Within the TNBC subgroup, no absolute increase in CNAs was found in the LNMs compared to their primary tumours, suggesting that increased genomic instability does not lead to more CNAs in LNMs. Our findings suggest a high clonal relationship between primary breast tumours and its LNMs, at least prior to treatment, and support the use of primary tumour characteristics to guide adjuvant systemic chemotherapy in breast cancer patients.
Collapse
Affiliation(s)
- Marieke A. Vollebergh
- Division of Molecular Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Division of Medical Oncology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Christiaan Klijn
- Division of Molecular Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Philip C. Schouten
- Division of Molecular Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Department of Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Danielle Israeli
- Department of Pathology, Vrije Universiteit University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, Vrije Universiteit University Medical Center, Amsterdam, the Netherlands
| | - Lodewyk F.A. Wessels
- Department of Bioinformatics and Statistics, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Sabine C. Linn
- Division of Molecular Pathology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Division of Medical Oncology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
33
|
Vollebergh MA, Lips EH, Nederlof PM, Wessels LFA, Wesseling J, Vd Vijver MJ, de Vries EGE, van Tinteren H, Jonkers J, Hauptmann M, Rodenhuis S, Linn SC. Genomic patterns resembling BRCA1- and BRCA2-mutated breast cancers predict benefit of intensified carboplatin-based chemotherapy. Breast Cancer Res 2014; 16:R47. [PMID: 24887359 PMCID: PMC4076636 DOI: 10.1186/bcr3655] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 05/02/2014] [Indexed: 12/12/2022] Open
Abstract
Introduction BRCA-mutated breast cancer cells lack the DNA-repair mechanism homologous recombination that is required for error-free DNA double-strand break (DSB) repair. Homologous recombination deficiency (HRD) may cause hypersensitivity to DNA DSB-inducing agents, such as bifunctional alkylating agents and platinum salts. HRD can be caused by BRCA mutations, and by other mechanisms. To identify HRD, studies have focused on triple-negative (TN) breast cancers as these resemble BRCA1-mutated breast cancer closely and might also share this hypersensitivity. However, ways to identify HRD in non-BRCA-mutated, estrogen receptor (ER)-positive breast cancers have remained elusive. The current study provides evidence that genomic patterns resembling BRCA1- or BRCA2-mutated breast cancers can identify breast cancer patients with TN as well as ER-positive, HER2-negative tumors that are sensitive to intensified, DSB-inducing chemotherapy. Methods Array comparative genomic hybridization (aCGH) was used to classify breast cancers. Patients with tumors with similar aCGH patterns as BRCA1- and/or BRCA2-mutated breast cancers were defined as having a BRCA-likeCGH status, others as non-BCRA-likeCGH. Stage-III patients (n = 249) had participated in a randomized controlled trial of adjuvant high-dose (HD) cyclophosphamide-thiotepa-carboplatin (CTC) versus 5-fluorouracil-epirubicin-cyclophosphamide (FE90C) chemotherapy. Results Among patients with BRCA-likeCGH tumors (81/249, 32%), a significant benefit of HD-CTC compared to FE90C was observed regarding overall survival (adjusted hazard ratio 0.19, 95% CI: 0.08 to 0.48) that was not seen for patients with non-BRCA-likeCGH tumors (adjusted hazard ratio 0.90, 95% CI: 0.53 to 1.54) (P = 0.004). Half of all BRCA-likeCGH tumors were ER-positive. Conclusions Distinct aCGH patterns differentiated between HER2-negative patients with a markedly improved outcome after adjuvant treatment with an intensified DNA-DSB-inducing regimen (BRCA-likeCGH patients) and those without benefit (non-BRCA-likeCGH patients).
Collapse
|
34
|
Systemically treated breast cancer patients and controls: an evaluation of the presence of noncredible performance. J Int Neuropsychol Soc 2014; 20:357-69. [PMID: 24607070 DOI: 10.1017/s1355617714000022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This study sought to define the frequency of noncredible performance in breast cancer patients before, during and after completion of systemic treatment, as well as predictors of noncredible performance. We examined six datasets investigating the cognitive effects of chemotherapy and/or endocrine therapy. Embedded performance validity test (PVT) measures were identified and used to evaluate the datasets. One dataset included a standalone PVT. Possible noncredible performance was operationally defined as performance below criterion on three or more PVT indices. This was undertaken as cancer patients have been observed clinically to fail PVTs both in the context of external gain and independent of such motivators. A total of 534 breast cancer patients and 214 healthy controls were included in the analysis. Percentages of patients performing below cutoff on one or more PVT varied from 0% to 21.2%. Only 1 patient met the criterion of noncredible performance. Calculation of post-test probabilities indicated a more than 90% chance to detect noncredible performance. There is no evidence to suggest noncredible performance in breast cancer patients and healthy controls who choose to participate in research studies examining cognitive function. Thus, the observational data showing that non-central nervous system (CNS) cancer and therapies not targeting the CNS can have untoward effects on cognitive function are unlikely to be due to noncredible performance.
Collapse
|
35
|
Recchia F, Candeloro G, Cesta A, DI Staso M, Bonfili P, Gravina GL, DI Cesare E, Necozione S, Rea S. Anthracycline-based induction chemotherapy followed by concurrent cyclophosphamide, methotrexate and 5-fluorouracil and radiation therapy in surgically resected axillary node-positive breast cancer. Mol Clin Oncol 2014; 2:473-478. [PMID: 24772320 DOI: 10.3892/mco.2014.269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/14/2014] [Indexed: 01/23/2023] Open
Abstract
The present study aimed to determine the toxicity and efficacy of 4 courses of anthracyclines-taxane (AT) chemotherapy followed by radiation therapy (XRT) concurrent with cyclophosphamide, methotrexate and 5-fluorouracil (CMF) in surgically resected axillary node-positive (N+) breast cancer. A total of 200 women with N+ breast cancer were treated with adriamycin and docetaxel followed by XRT concurrent with six courses of CMF. Two courses of dose-dense chemotherapy with ifosfamide, carboplatin and etoposide, supported by pegfilgrastim, were administered to patients with >5 histologically confirmed axillary lymph node metastases and patients with triple-negative disease. Additional treatments included 1 year of trastuzumab in human epidermal growth factor receptor 2-positive patients, 5 years of a luteinizing hormone-releasing hormone analogue in premenopausal women and 5 years of an aromatase inhibitor (AI) in estrogen receptor-positive (ER+) patients. The mean number of positive axillary lymph nodes was 4.4 (range, 2-37), 52% of the patients were premenopausal, 74% were ER+ and 26% had triple-negative disease. After a median follow-up of 73 months, grade 2 and 3 hematological toxicity was observed in 20% of the patients. The 10-year disease-free survival (DFS) and overall survival (OS) rates were 73 and 77%, respectively. There was no significant difference in DFS between ER+ and estrogen receptor-negative (ER-) patients (P>0.05), whereas the OS was better in ER+ vs. ER- patients (P<0.05) and in premenopausal vs. postmenopausal patients (P<0.005). In conclusion, induction AT concurrent CMF and XRT and dose-dense chemotherapy followed by AI in N+ high-risk breast cancer was associated with a low level of systemic and late cardiac toxicity and excellent local control, DFS and OS.
Collapse
Affiliation(s)
- Francesco Recchia
- Department of Medical Oncology, Civilian Hospital, 67051 Avezzano, L'Aquila, Italy ; ; Carlo Ferri Foundation, 00013 Monterotondo, Rome; University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Giampiero Candeloro
- Department of Medical Oncology, Civilian Hospital, 67051 Avezzano, L'Aquila, Italy
| | - Alisia Cesta
- Department of Medical Oncology, Civilian Hospital, 67051 Avezzano, L'Aquila, Italy
| | - Mario DI Staso
- Laboratory of Radiobiology and Division of Radiotherapy, Department of Applied Clinical and Biotechnological Sciences; University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Pierluigi Bonfili
- Laboratory of Radiobiology and Division of Radiotherapy, Department of Applied Clinical and Biotechnological Sciences; University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Laboratory of Radiobiology and Division of Radiotherapy, Department of Applied Clinical and Biotechnological Sciences; University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Ernesto DI Cesare
- Laboratory of Radiobiology and Division of Radiotherapy, Department of Applied Clinical and Biotechnological Sciences; University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Stefano Necozione
- Departments of Clinical Epidemiology, University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| | - Silvio Rea
- Surgical Oncology, University of L'Aquila, 67100 L'Aquila, L'Aquila, Italy
| |
Collapse
|
36
|
High Dose Thiotepa in Patients with Relapsed or Refractory Osteosarcomas: Experience of the SFCE Group. Sarcoma 2014; 2014:475067. [PMID: 24672280 PMCID: PMC3941142 DOI: 10.1155/2014/475067] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/15/2013] [Indexed: 11/24/2022] Open
Abstract
Introduction. Osteosarcoma relapse has a poor prognosis, with less than 25% survival at 5 years. We describe the experience of the French Society of Paediatric Oncology (SFCE) with high dose (HD) thiotepa and autologous stem cell transplantation (ASCT) in 45 children with relapsed osteosarcoma. Patients and Methods. Between 1992 and 2004, 53 patients received HD thiotepa (900 mg/m2) followed by ASCT in 6 centres. Eight patients were excluded from analysis, and we retrospectively reviewed the clinical radiological and anatomopathological patterns of the 45 remaining patients. Results. Sixteen girls and 29 boys (median age, 15.9 years) received HD thiotepa after initial progression of metastatic disease (2), first relapse (26), and second or third relapse (17). We report 12 radiological partial responses and 9 of 31 histological complete responses. Thirty-two patients experienced further relapses, and 13 continued in complete remission after surgical resection of the residual disease. Three-year overall survival was 40%, and 3-year progression-free survival was 24%. Delay of relapse (+/− 2 years from diagnosis) was a prognostic factor (P = 0.011). No acute toxic serious adverse event occurred. Conclusion. The use of HD thiotepa and ASCT is feasible in patients with relapsed osteosarcoma. A randomized study for recurrent osteosarcoma between standard salvage chemotherapy and high dose thiotepa with stem cell rescue is ongoing.
Collapse
|
37
|
A Case-Control Comparison of Retracted and Non-Retracted Clinical Trials: Can Retraction Be Predicted? PUBLICATIONS 2014. [DOI: 10.3390/publications2010027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
38
|
Henderson IC, Bhatia V. Nab-paclitaxel for breast cancer: a new formulation with an improved safety profile and greater efficacy. Expert Rev Anticancer Ther 2014; 7:919-43. [PMID: 17627452 DOI: 10.1586/14737140.7.7.919] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Taxanes, paclitaxel and docetaxel, are among the most effective agents used to treat breast cancer. Nab-paclitaxel (ABI-007, Abraxane) is paclitaxel encapsulated in albumin. This differs from the more conventional formulation which uses cremophor to increase the solubility of paclitaxel (CrEL-paclitaxel). In a randomized trial that formed the basis of its regulatory approval in the USA, 3-weekly nab-paclitaxel induced a higher response rate and longer time to progression than CrEL-paclitaxel in patients with metastatic breast cancer. Except for grade 3 sensory neuropathy, nab-paclitaxel was also safer. An interim analysis from a more recent randomized Phase II trial suggests that weekly nab-paclitaxel is more effective and safer than either 3-weekly nab-paclitaxel or 3-weekly docetaxel. The superior efficacy of nab-paclitaxel is presumably due to the improved safety profile, which allows for the administration of higher doses, a greater proportion of which actually reaches the tumor. Observations on the development of nab-paclitaxel have important implications for our understanding of dose response in the use of cytotoxic drugs to treat all forms of cancer. Although it is not yet clear whether nab-paclitaxel can be routinely substituted for CrEL-paclitaxel or docetaxel in breast cancer treatment regimens, it seems highly likely that this will occur within the next 5 years.
Collapse
Affiliation(s)
- I Craig Henderson
- Adjunct Professor of Medicine, University of California, San Francisco, UCSF Comprehensive Cancer Center, San Francisco, CA 94143, USA.
| | | |
Collapse
|
39
|
Pedrazzoli P, Martinelli G, Gianni AM, Da Prada GA, Ballestrero A, Rosti G, Frassineti GL, Aieta M, Secondino S, Cinieri S, Fedele R, Bengala C, Bregni M, Grasso D, De Giorgi U, Lanza F, Castagna L, Bruno B, Martino M. Adjuvant high-dose chemotherapy with autologous hematopoietic stem cell support for high-risk primary breast cancer: results from the Italian national registry. Biol Blood Marrow Transplant 2013; 20:501-6. [PMID: 24374214 DOI: 10.1016/j.bbmt.2013.12.569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 12/23/2013] [Indexed: 01/07/2023]
Abstract
The efficacy of high-dose chemotherapy (HDC) and autologous hemopoietic progenitor cell transplantation (AHPCT) for breast cancer (BC) patients has been an area of intense controversy among the medical oncology community. The aim of this study was to assess toxicity and efficacy of this procedure in a large cohort of high-risk primary BC patients who underwent AHPCT in Italy. A total of 1183 patients receiving HDC for high-risk BC (HRBC) (>3 positive nodes) were identified in the Italian registry. The median age was 46 years, 62% of patients were premenopausal at treatment, 60.1% had endocrine-responsive tumors, and 20.7% had a human epidermal growth factor receptor 2 (HER2)-positive tumor. The median number of positive lymph nodes (LN) at surgery was 15, with 71.5% of patients having ≥ 10 positive nodes. Seventy-three percent received an alkylating agent-based HDC as a single procedure, whereas 27% received epirubicin or mitoxantrone-containing HDC, usually within a multitransplantation program. The source of stem cells was peripheral blood in the vast majority of patients. Transplantation-related mortality was .8%, whereas late cardiac and secondary tumor-related mortality were around 1%, overall. With a median follow-up of 79 months, median disease-free and overall survival (OS) in the entire population were 101 and 134 months, respectively. Subgroup analysis demonstrated that OS was significantly better in patients with endocrine-responsive tumors and in patients receiving multiple transplantation procedures. HER2 status did not affect survival probability. The size of the primary tumor and number of involved LN negatively affected OS. Adjuvant HDC with AHPCT has a low mortality rate and provides impressive long-term survival rates in patients with high-risk primary BC. Our results suggest that this treatment modality should be proposed in selected HRBC patients and further investigated in clinical trials.
Collapse
Affiliation(s)
- Paolo Pedrazzoli
- Medical Oncology, IRCCS Foundation, San Matteo Hospital, Pavia, Italy
| | | | | | | | | | | | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | - Michele Aieta
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | | | - Saverio Cinieri
- Medical Oncology, European Institute of Oncology, Milan, Italy
| | - Roberta Fedele
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera "BMM", Reggio Calabria, Italy
| | | | - Marco Bregni
- Hematology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Donatella Grasso
- Medical Oncology, IRCCS Foundation, San Matteo Hospital, Pavia, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | - Francesco Lanza
- Section of Hematology and Bone Marrow Transplant Unit, Cremona, Italy
| | - Luca Castagna
- Hematology Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Barbara Bruno
- National Registry GITMO & Data Managing, Ospedale San Martino, Genova, Italy
| | - Massimo Martino
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera "BMM", Reggio Calabria, Italy.
| | | |
Collapse
|
40
|
Pedrazzoli P, Tarenzi E, Tullio C, Colosini G, Siena S. High Dose Chemotherapy and Hematopoietic Progenitor Cell Transplantation for Breast Cancer. J Chemother 2013; 16 Suppl 4:108-11. [PMID: 15688624 DOI: 10.1179/joc.2004.16.supplement-1.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Nowhere has there been more controversy in recent years than in the use of high dose chemotherapy (HDC) with autologous hematopoietic stem cell transplantation for breast cancer, both in the adjuvant setting and for advanced disease. Authors review and comment on the data from the studies so far reported and try to indicate what will be next in this field. They also discuss what may be the attitude to take in our everyday clinical practice, taking into account the availability of new chemotherapeutic agents and targeted therapies.
Collapse
Affiliation(s)
- P Pedrazzoli
- SC Oncologia Medica Falck, Dipartimento Oncologico, Ospedale Niguarda Ca' Granda, 1-20162 Milano, Italy.
| | | | | | | | | |
Collapse
|
41
|
Breast cancer follow-up strategies in randomized phase III adjuvant clinical trials: a systematic review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:89. [PMID: 24438135 PMCID: PMC3828573 DOI: 10.1186/1756-9966-32-89] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/07/2013] [Indexed: 01/02/2023]
Abstract
The effectiveness of different breast cancer follow-up procedures to decrease breast cancer mortality are still an object of debate, even if intensive follow-up by imaging modalities is not recommended by international guidelines since 1997. We conducted a systematic review of surveillance procedures utilized, in the last ten years, in phase III randomized trials (RCTs) of adjuvant treatments in early stage breast cancer with disease free survival as primary endpoint of the study, in order to verify if a similar variance exists in the scientific world. Follow-up modalities were reported in 66 RCTs, and among them, minimal and intensive approaches were equally represented, each being followed by 33 (50%) trials. The minimal surveillance regimen is preferred by international and North American RCTs (P = 0.001) and by trials involving more than one country (P = 0.004), with no relationship with the number of participating centers (P = 0.173), with pharmaceutical industry sponsorship (P = 0.80) and with trials enrolling > 1000 patients (P = 0.14). At multivariate regression analysis, only geographic location of the trial was predictive for a distinct follow-up methodology (P = 0.008): Western European (P = 0.004) and East Asian studies (P = 0.010) use intensive follow-up procedures with a significantly higher frequency than international RCTs, while no differences have been detected between North American and international RCTs. Stratifying the studies according to the date of beginning of patients enrollment, before or after 1998, in more recent RCTs the minimal approach is more frequently followed by international and North American RCTs (P = 0.01), by trials involving more than one country (P = 0.01) and with more than 50 participating centers (P = 0.02). It would be highly desirable that in the near future breast cancer follow-up procedures will be homogeneous in RCTs and everyday clinical settings.
Collapse
|
42
|
Schouten PC, van Dyk E, Braaf LM, Mulder L, Lips EH, de Ronde JJ, Holtman L, Wesseling J, Hauptmann M, Wessels LFA, Linn SC, Nederlof PM. Platform comparisons for identification of breast cancers with a BRCA-like copy number profile. Breast Cancer Res Treat 2013; 139:317-27. [PMID: 23670131 DOI: 10.1007/s10549-013-2558-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 04/29/2013] [Indexed: 12/28/2022]
Abstract
Previously, we employed bacterial artificial chromosome (BAC) array comparative genomic hybridization (aCGH) profiles from BRCA1 and -2 mutation carriers and sporadic tumours to construct classifiers that identify tumour samples most likely to harbour BRCA1 and -2 mutations, designated 'BRCA1 and -2-like' tumours, respectively. The classifiers are used in clinical genetics to evaluate unclassified variants, and patients for which no good quality germline DNA is available. Furthermore, we have shown that breast cancer patients with BRCA-like tumour aCGH profiles benefit substantially from platinum-based chemotherapy, potentially due to their inability to repair DNA double strand breaks (DSB), providing a further important clinical application for the classifiers. The BAC array technology has been replaced with oligonucleotide arrays. To continue clinical use of existing classifiers, we mapped oligonucleotide aCGH data to the BAC domain, such that the oligonucleotide profiles can be employed as in the BAC classifier. We demonstrate that segmented profiles derived from oligonucleotide aCGH show high correlation with BAC aCGH profiles. Furthermore, we trained a support vector machine score to objectify aCGH profile quality. Using the mapped oligonucleotide aCGH data, we show equivalence in classification of biologically relevant cases between BAC and oligonucleotide data. Furthermore, the predicted benefit of DSB inducing chemotherapy due to a homologous recombination defect is retained. We conclude that oligonucleotide aCGH data can be mapped to and used in the previously developed and validated BAC aCGH classifiers. Our findings suggest that it is possible to map copy number data from any other technology in a similar way.
Collapse
Affiliation(s)
- Philip C Schouten
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
High-dose chemotherapy followed by autologous stem cell transplantation as a first-line therapy for high-risk primary breast cancer: a meta-analysis. PLoS One 2012; 7:e33388. [PMID: 22428041 PMCID: PMC3299795 DOI: 10.1371/journal.pone.0033388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 02/13/2012] [Indexed: 11/28/2022] Open
Abstract
Background and Objectives Several trials have generated conflicting results about the results of high-dose chemotherapy followed by autologous stem cell transplantation (HDCT) for primary breast cancer. This meta-analysis summarizes the available evidence from all suitable studies. Design and Methods Prospective, randomized trials with HDCT as a first-line therapy for primary breast cancer were included in this meta-analysis. The primary outcome of interest for our analysis was survival (disease-free survival and overall survival); secondary endpoints included treatment-related mortality (TRM) and second (non-breast) cancers. We used a median age of 47, a PR positive rate of 50% and a premenopausal rate of 70% as cutoff values to complete the subgroup analyses, which were pre-planned according to the prepared protocol. Results Fourteen trials with 5747 patients were eligible for the meta-analysis. Compared with non-HDCT, non-significant second (non-breast) cancers (RR = 1.28; 95% CI = 0.82–1.98) and higher TRM (RR = 3.42; 95% CI = 1.32–8.86) were associated with HDCT for primary breast cancer. A significant DFS benefit of HDCT was documented (HR = 0.89; 95% CI = 0.79–0.99). No difference in OS (overall survival) was found when the studies were pooled (HR = 0.91; 95% CI = 0.82–1.00, p = 0.062). In subgroup analysis, age and hormone receptor status had a significant interaction with prolonged DFS and OS. Conclusions HDCT has a benefit on DFS and OS compared to SDC in some special patients with high-risk primary breast cancer.
Collapse
|
44
|
Rottenberg S, Vollebergh MA, de Hoon B, de Ronde J, Schouten PC, Kersbergen A, Zander SAL, Pajic M, Jaspers JE, Jonkers M, Lodén M, Sol W, van der Burg E, Wesseling J, Gillet JP, Gottesman MM, Gribnau J, Wessels L, Linn SC, Jonkers J, Borst P. Impact of intertumoral heterogeneity on predicting chemotherapy response of BRCA1-deficient mammary tumors. Cancer Res 2012; 72:2350-61. [PMID: 22396490 DOI: 10.1158/0008-5472.can-11-4201] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The lack of markers to predict chemotherapy responses in patients poses a major handicap in cancer treatment. We searched for gene expression patterns that correlate with docetaxel or cisplatin response in a mouse model for breast cancer associated with BRCA1 deficiency. Array-based expression profiling did not identify a single marker gene predicting docetaxel response, despite an increase in Abcb1 (P-glycoprotein) expression that was sufficient to explain resistance in several poor responders. Intertumoral heterogeneity explained the inability to identify a predictive gene expression signature for docetaxel. To address this problem, we used a novel algorithm designed to detect differential gene expression in a subgroup of the poor responders that could identify tumors with increased Abcb1 transcript levels. In contrast, standard analytical tools, such as significance analysis of microarrays, detected a marker only if it correlated with response in a substantial fraction of tumors. For example, low expression of the Xist gene correlated with cisplatin hypersensitivity in most tumors, and it also predicted long recurrence-free survival of HER2-negative, stage III breast cancer patients treated with intensive platinum-based chemotherapy. Our findings may prove useful for selecting patients with high-risk breast cancer who could benefit from platinum-based therapy.
Collapse
Affiliation(s)
- Sven Rottenberg
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Companion diagnostics: changing patient management. Ecancermedicalscience 2012; 6:244. [PMID: 22423251 PMCID: PMC3298409 DOI: 10.3332/ecancer.2012.244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Indexed: 11/06/2022] Open
Abstract
At the European Multidisciplinary Cancer Congress (EMCC), held in Stockholm in September 2011, a recurring theme in many of the workshops was personalised medicine, including the latest developments in prognostic and predictive biomarkers. Such markers, it is hoped, will enable clinicians to use available resources to best effects-by offering treatments to only those patients most likely to benefit, or by avoiding treatments that are likely to cause toxicities with limited benefit. The emergence of novel diagnostic tools that can distinguish subsets of patients with different response to treatment is likely to result in a paradigm shift in the way in which we manage cancer in the future. This report focuses on some of the key developments and challenges in providing a truly individualised approach to therapy, as presented at EMCC 2011.
Collapse
|
46
|
Coates AS, Colleoni M, Goldhirsch A. Is adjuvant chemotherapy useful for women with luminal a breast cancer? J Clin Oncol 2012; 30:1260-3. [PMID: 22355052 DOI: 10.1200/jco.2011.37.7879] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Alan S Coates
- International Breast Cancer Study Group and University of Sydney, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
47
|
Quantitative copy number analysis by Multiplex Ligation-dependent Probe Amplification (MLPA) of BRCA1-associated breast cancer regions identifies BRCAness. Breast Cancer Res 2011; 13:R107. [PMID: 22032731 PMCID: PMC3262220 DOI: 10.1186/bcr3049] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/12/2011] [Accepted: 10/27/2011] [Indexed: 12/21/2022] Open
Abstract
Introduction Our group has previously employed array Comparative Genomic Hybridization (aCGH) to assess the genomic patterns of BRCA1-mutated breast cancers. We have shown that the so-called BRCA1-likeaCGH profile is also present in about half of all triple-negative sporadic breast cancers and is predictive for benefit from intensified alkylating chemotherapy. As aCGH is a rather complex method, we translated the BRCA1aCGH profile to a Multiplex Ligation-dependent Probe Amplification (MLPA) assay, to identify both BRCA1-mutated breast cancers and sporadic cases with a BRCA1-likeaCGH profile. Methods The most important genomic regions of the original aCGH based classifier (3q22-27, 5q12-14, 6p23-22, 12p13, 12q21-23, 13q31-34) were mapped to a set of 34 MLPA probes. The training set consisted of 39 BRCA1-likeaCGH breast cancers and 45 non-BRCA1-likeaCGH breast cancers, which had previously been analyzed by aCGH. The BRCA1-likeaCGH group consisted of germline BRCA1-mutated cases and sporadic tumours with low BRCA1 gene expression and/or BRCA1 promoter methylation. We trained a shrunken centroids classifier on the training set and validation was performed on an independent test set of 40 BRCA1-likeaCGH breast cancers and 32 non-BRCA1-likeaCGH breast cancer tumours. In addition, we validated the set prospectively on 69 new triple-negative tumours. Results BRCAness in the training set of 84 tumours could accurately be predicted by prediction analysis of microarrays (PAM) (accuracy 94%). Application of this classifier on the independent validation set correctly predicted BRCA-like status of 62 out of 72 breast tumours (86%). Sensitivity and specificity were 85% and 87%, respectively. When the MLPA-test was subsequently applied to 46 breast tumour samples from a randomized clinical trial, the same survival benefit for BRCA1-like tumours associated with intensified alkylating chemotherapy was shown as was previously reported using the aCGH assay. Conclusions Since the MLPA assay can identify BRCA1-deficient breast cancer patients, this method could be applied both for clinical genetic testing and as a predictor of treatment benefit. BRCA1-like tumours are highly sensitive to chemotherapy with DNA damaging agents, and most likely to poly ADP ribose polymerase (PARP)-inhibitors. The MLPA assay is rapid and robust, can easily be multiplexed, and works well with DNA derived from paraffin-embedded tissues.
Collapse
|
48
|
Gast MCW, Zapatka M, van Tinteren H, Bontenbal M, Span PN, Tjan-Heijnen VCG, Knol JC, Jimenez CR, Schellens JHM, Beijnen JH. Postoperative serum proteomic profiles may predict recurrence-free survival in high-risk primary breast cancer. J Cancer Res Clin Oncol 2011; 137:1773-83. [PMID: 21913038 PMCID: PMC3205273 DOI: 10.1007/s00432-011-1055-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 08/30/2011] [Indexed: 11/30/2022]
Abstract
PURPOSE Better breast cancer prognostication may improve selection of patients for adjuvant therapy. We conducted a retrospective longitudinal study in which we investigated sera of high-risk primary breast cancer patients, to search for proteins predictive of recurrence-free survival. METHODS Sera of 82 breast cancer patients obtained after surgery, but prior to the administration of adjuvant therapy, were fractionated using anion-exchange chromatography, to facilitate the detection of the low-abundant serum peptides. Selected fractions were subsequently analysed by surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS), and the resulting protein profiles were searched for prognostic markers by appropriate bioinformatics tools. RESULTS Four peak clusters (i.e. m/z 3073, m/z 3274, m/z 4405 and m/z 7973) were found to bear significant prognostic value (P ≤ 0.01). The m/z 3274 candidate marker was structurally identified as inter-alpha-trypsin inhibitor heavy chain 4 fragment(658-688) in serum. Except for the m/z 7973 peak cluster, these peaks remained independently associated with recurrence-free survival upon multivariate Cox regression analysis, including clinical parameters of known prognostic value in this study population. CONCLUSION Investigation of the postoperative serum proteome by, e.g., anion-exchange fractionation followed by SELDI-TOF MS analysis is promising for the detection of novel prognostic factors. However, regarding the rather limited study population, validation of these results by analysis of independent study populations is warranted to assess the true clinical applicability of discovered prognostic markers. In addition, structural identification of the other markers will aid in elucidation of their role in breast cancer prognosis, as well as enable development of absolute quantitative assays.
Collapse
Affiliation(s)
- Marie-Christine W Gast
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Slotervaart Hospital, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Berry DA, Ueno NT, Johnson MM, Lei X, Caputo J, Rodenhuis S, Peters WP, Leonard RC, Barlow WE, Tallman MS, Bergh J, Nitz UA, Gianni AM, Basser RL, Zander AR, Coombes RC, Roché H, Tokuda Y, de Vries EGE, Hortobagyi GN, Crown JP, Pedrazzoli P, Bregni M, Demirer T. High-dose chemotherapy with autologous stem-cell support as adjuvant therapy in breast cancer: overview of 15 randomized trials. J Clin Oncol 2011; 29:3214-23. [PMID: 21768471 DOI: 10.1200/jco.2010.32.5910] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Adjuvant high-dose chemotherapy (HDC) with autologous hematopoietic stem-cell transplantation (AHST) for high-risk primary breast cancer has not been shown to prolong survival. Individual trials have had limited power to show overall benefit or benefits within subsets. METHODS We assembled individual patient data from 15 randomized trials that compared HDC versus control therapy without stem-cell support. Prospectively defined primary end points were relapse-free survival (RFS) and overall survival (OS). We compared the effect of HDC versus control by using log-rank tests and proportional hazards regression, and we adjusted for clinically relevant covariates. Subset analyses were by age, number of positive lymph nodes, tumor size, histology, hormone receptor (HmR) status, and human epidermal growth factor receptor 2 (HER2) status. RESULTS Of 6,210 total patients (n = 3,118, HDC; n = 3,092 control), the median age was 46 years; 69% were premenopausal, 29% were postmenopausal, and 2% were unknown menopausal status; 49.5% were HmR positive; 33.5% were HmR negative, and 17% were unknown HmR status. The median follow-up was 6 years. After analysis was adjusted for covariates, HDC was found to prolong relapse-free survival (RFS; hazard ratio [HR], 0.87; 95% CI, 0.81 to 0.93; P < .001) but not overall survival (OS; HR, 0.94; 95% CI, 0.87 to 1.02; P = .13). For OS, no covariates had statistically significant interactions with treatment effect, and no subsets evinced a significant effect of HDC. Younger patients had a significantly better RFS on HDC than did older patients. CONCLUSION Adjuvant HDC with AHST prolonged RFS in high-risk primary breast cancer compared with control, but this did not translate into a significant OS benefit. Whether HDC benefits patients in the context of targeted therapies is unknown.
Collapse
Affiliation(s)
- Donald A Berry
- Division of Quantitative Sciences, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77230-1402, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Vollebergh MA, Lips EH, Nederlof PM, Wessels LFA, Schmidt MK, van Beers EH, Cornelissen S, Holtkamp M, Froklage FE, de Vries EGE, Schrama JG, Wesseling J, van de Vijver MJ, van Tinteren H, de Bruin M, Hauptmann M, Rodenhuis S, Linn SC. An aCGH classifier derived from BRCA1-mutated breast cancer and benefit of high-dose platinum-based chemotherapy in HER2-negative breast cancer patients. Ann Oncol 2011; 22:1561-1570. [PMID: 21135055 PMCID: PMC3121967 DOI: 10.1093/annonc/mdq624] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/07/2010] [Accepted: 09/14/2010] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breast cancer cells deficient for BRCA1 are hypersensitive to agents inducing DNA double-strand breaks (DSB), such as bifunctional alkylators and platinum agents. Earlier, we had developed a comparative genomic hybridisation (CGH) classifier based on BRCA1-mutated breast cancers. We hypothesised that this BRCA1-like(CGH) classifier could also detect loss of function of BRCA1 due to other causes besides mutations and, consequently, might predict sensitivity to DSB-inducing agents. PATIENTS AND METHODS We evaluated this classifier in stage III breast cancer patients, who had been randomly assigned between adjuvant high-dose platinum-based (HD-PB) chemotherapy, a DSB-inducing regimen, and conventional anthracycline-based chemotherapy. Additionally, we assessed BRCA1 loss through mutation or promoter methylation and immunohistochemical basal-like status in the triple-negative subgroup (TN subgroup). RESULTS We observed greater benefit from HD-PB chemotherapy versus conventional chemotherapy among patients with BRCA1-like(CGH) tumours [41/230 = 18%, multivariate hazard ratio (HR) = 0.12, 95% confidence interval (CI) 0.04-0.43] compared with patients with non-BRCA1-like(CGH) tumours (189/230 = 82%, HR = 0.78, 95% CI 0.50-1.20), with a significant difference (test for interaction P = 0.006). Similar results were obtained for overall survival (P interaction = 0.04) and when analyses were restricted to the TN subgroup. Sixty-three percent (20/32) of assessable BRCA1-like(CGH) tumours harboured either a BRCA1 mutation (n = 8) or BRCA1 methylation (n = 12). CONCLUSION BRCA1 loss as assessed by CGH analysis can identify patients with substantially improved outcome after adjuvant DSB-inducing chemotherapy when compared with standard anthracycline-based chemotherapy in our series.
Collapse
Affiliation(s)
- M A Vollebergh
- Division of Molecular Biology; Division of Medical Oncology
| | - E H Lips
- Division of Experimental Therapy
| | - P M Nederlof
- Division of Experimental Therapy; Division of Molecular Pathology
| | - L F A Wessels
- Department of Bioinformatics and Statistics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam; Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft
| | - M K Schmidt
- Division of Experimental Therapy; Department of Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | | | | | | | | | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen
| | | | | | - M J van de Vijver
- Department of Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam; Department of Pathology, Academic Medical Center
| | - H van Tinteren
- Department of Biometrics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - M Hauptmann
- Department of Bioinformatics and Statistics, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | | | - S C Linn
- Division of Molecular Biology; Division of Medical Oncology.
| |
Collapse
|