751
|
Gianni L, Pienkowski T, Im YH, Roman L, Tseng LM, Liu MC, Lluch A, Staroslawska E, de la Haba-Rodriguez J, Im SA, Pedrini JL, Poirier B, Morandi P, Semiglazov V, Srimuninnimit V, Bianchi G, Szado T, Ratnayake J, Ross G, Valagussa P. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2011; 13:25-32. [PMID: 22153890 DOI: 10.1016/s1470-2045(11)70336-9] [Citation(s) in RCA: 1647] [Impact Index Per Article: 117.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Studies with pertuzumab, a novel anti-HER2 antibody, show improved efficacy when combined with the established HER2-directed antibody trastuzumab in breast cancer therapy. We investigated the combination of pertuzumab or trastuzumab, or both, with docetaxel and the combination of pertuzumab and trastuzumab without chemotherapy in the neoadjuvant setting. METHODS In this multicentre, open-label, phase 2 study, treatment-naive women with HER2-positive breast cancer were randomly assigned (1:1:1:1) centrally and stratified by operable, locally advanced, and inflammatory breast cancer, and by hormone receptor expression to receive four neoadjuvant cycles of: trastuzumab (8 mg/kg loading dose, followed by 6 mg/kg every 3 weeks) plus docetaxel (75 mg/m(2), escalating, if tolerated, to 100 mg/m(2) every 3 weeks; group A) or pertuzumab (loading dose 840 mg, followed by 420 mg every 3 weeks) and trastuzumab plus docetaxel (group B) or pertuzumab and trastuzumab (group C) or pertuzumab plus docetaxel (group D). The primary endpoint, examined in the intention-to-treat population, was pathological complete response in the breast. Neither patients nor investigators were masked to treatment. This study is registered with ClinicalTrials.gov, number NCT00545688. FINDINGS Of 417 eligible patients, 107 were randomly assigned to group A, 107 to group B, 107 to group C, and 96 to group D. Patients given pertuzumab and trastuzumab plus docetaxel (group B) had a significantly improved pathological complete response rate (49 of 107 patients; 45·8% [95% CI 36·1-55·7]) compared with those given trastuzumab plus docetaxel (group A; 31 of 107; 29·0% [20·6-38·5]; p=0·0141). 23 of 96 (24·0% [15·8-33·7]) women given pertuzumab plus docetaxel (group D) had a pathological complete response, as did 18 of 107 (16·8% [10·3-25·3]) given pertuzumab and trastuzumab (group C). The most common adverse events of grade 3 or higher were neutropenia (61 of 107 women in group A, 48 of 107 in group B, one of 108 in group C, and 52 of 94 in group D), febrile neutropenia (eight, nine, none, and seven, respectively), and leucopenia (13, five, none, and seven, respectively). The number of serious adverse events was similar in groups A, B, and D (15-20 serious adverse events per group in 10-17% of patients) but lower in group C (four serious adverse events in 4% of patients). INTERPRETATION Patients given pertuzumab and trastuzumab plus docetaxel (group B) had a significantly improved pathological complete response rate compared with those given trastuzumab plus docetaxel, without substantial differences in tolerability. Pertuzumab and trastuzumab without chemotherapy eradicated tumours in a proportion of women and showed a favourable safety profile. These findings justify further exploration in adjuvant trials and support the neoadjuvant approach for accelerating drug assessment in early breast cancer. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Luca Gianni
- Oncologia Medica, San Raffaele Cancer Centre, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
752
|
Hsu JT, Chen TC, Tseng JH, Chiu CT, Liu KH, Yeh CN, Hwang TL, Jan YY, Yeh TS. Impact of HER-2 overexpression/amplification on the prognosis of gastric cancer patients undergoing resection: a single-center study of 1,036 patients. Oncologist 2011; 16:1706-1713. [PMID: 22143936 PMCID: PMC3248769 DOI: 10.1634/theoncologist.2011-0199] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/06/2011] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Opinions regarding the impact of human epidermal growth factor receptor (HER)-2 overexpression or HER-2 amplification on the prognosis of gastric cancer patients are mixed. The present study attempted to clarify this issue by investigating a large cohort of surgical patients. METHODS We investigated 1,036 gastric cancer patients undergoing curative-intent resection. Their surgical specimens were evaluated for HER-2 expression by immunohistochemistry (IHC), and those with HER-2 expression levels of 2+ were additionally subjected to fluorescence in situ hybridization (FISH). Data on demographic and clinicopathological features and relevant prognostic factors in these patients were analyzed. RESULTS HER-2 positivity was noted in 64 (6.1%) of 1,036 gastric cancer patients, including 46 patients whose HER-2 expression level was 3+ on IHC and 18 patients whose FISH results were positive. On univariate analysis, HER-2 positivity was more often associated with differentiated histology, intestinal type, and negative resection margins, whereas only differentiated histology was independently associated with HER-2 positivity in a logistic regression model. For stage I-IV gastric cancer, HER-2 was not a prognostic factor. In a subpopulation study, although HER-2 positivity emerged as a favorable prognostic factor for stage III-IV gastric cancer on univariate analysis, it failed to be an independent prognostic factor after multivariate adjustment. CONCLUSIONS The prevalence of HER-2 positivity, determined using standardized assays and scoring criteria in a large cohort of gastric cancer patients after resection, was 6.1%. HER-2 positivity was phenotypically associated with differentiated histology. HER-2 is not an independent prognostic factor for gastric cancer.
Collapse
Affiliation(s)
| | | | | | - Cheng-Tang Chiu
- Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
753
|
Abstract
The advent of HER2-directed therapies has significantly improved the outlook for patients with HER2-positive early stage breast cancer. However, a significant proportion of these patients still relapse and die of breast cancer. Trials to define, refine and optimize the use of the two approved HER2-targeted agents (trastuzumab and lapatinib) in patients with HER2-positive early stage breast cancer are ongoing. In addition, promising new approaches are being developed including monoclonal antibodies and small-molecule tyrosine kinase inhibitors targeting HER2 or other HER family members, antibodies linked to cytotoxic moieties or modified to improve their immunological function, immunostimulatory peptides, and targeting the PI3K and IGF-1R pathways. Improved understanding of the HER2 signaling pathway, its relationship with other signaling pathways and mechanisms of resistance has also led to the development of rational combination therapies and to a greater insight into treatment response in patients with HER2-positive breast cancer. Based on promising results with new agents in HER2-positive advanced-stage disease, a series of large trials in the adjuvant and neoadjuvant settings are planned or ongoing. This Review focuses on current treatment for patients with HER2-positive breast cancer and aims to update practicing clinicians on likely future developments in the treatment for this disease according to ongoing clinical trials and translational research.
Collapse
|
754
|
Hamid O, Schmidt H, Nissan A, Ridolfi L, Aamdal S, Hansson J, Guida M, Hyams DM, Gómez H, Bastholt L, Chasalow SD, Berman D. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med 2011; 9:204. [PMID: 22123319 PMCID: PMC3239318 DOI: 10.1186/1479-5876-9-204] [Citation(s) in RCA: 441] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 11/28/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Ipilimumab, a fully human monoclonal antibody that blocks cytotoxic T-lymphocyte antigen-4, has demonstrated an improvement in overall survival in two phase III trials of patients with advanced melanoma. The primary objective of the current trial was to prospectively explore candidate biomarkers from the tumor microenvironment for associations with clinical response to ipilimumab. METHODS In this randomized, double-blind, phase II biomarker study (ClinicalTrials.gov NCT00261365), 82 pretreated or treatment-naïve patients with unresectable stage III/IV melanoma were induced with 3 or 10 mg/kg ipilimumab every 3 weeks for 4 doses; at Week 24, patients could receive maintenance doses every 12 weeks. Efficacy was evaluated per modified World Health Organization response criteria and safety was assessed continuously. Candidate biomarkers were evaluated in tumor biopsies collected pretreatment and 24 to 72 hours after the second ipilimumab dose. Polymorphisms in immune-related genes were also evaluated. RESULTS Objective response rate, response patterns, and safety were consistent with previous trials of ipilimumab in melanoma. No associations between genetic polymorphisms and clinical activity were observed. Immunohistochemistry and histology on tumor biopsies revealed significant associations between clinical activity and high baseline expression of FoxP3 (p = 0.014) and indoleamine 2,3-dioxygenase (p = 0.012), and between clinical activity and increase in tumor-infiltrating lymphocytes (TILs) between baseline and 3 weeks after start of treatment (p = 0.005). Microarray analysis of mRNA from tumor samples taken pretreatment and post-treatment demonstrated significant increases in expression of several immune-related genes, and decreases in expression of genes implicated in cancer and melanoma. CONCLUSIONS Baseline expression of immune-related tumor biomarkers and a post-treatment increase in TILs may be positively associated with ipilimumab clinical activity. The observed pharmacodynamic changes in gene expression warrant further analysis to determine whether treatment-emergent changes in gene expression may be associated with clinical efficacy. Further studies are required to determine the predictive value of these and other potential biomarkers associated with clinical response to ipilimumab.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunohistochemistry
- Ipilimumab
- Male
- Melanoma/drug therapy
- Melanoma/genetics
- Melanoma/pathology
- Middle Aged
- Neoplasm Staging
- Polymorphism, Genetic
- Prospective Studies
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Young Adult
Collapse
Affiliation(s)
- Omid Hamid
- Melanoma Center, The Angeles Clinic and Research Institute, (2001 Santa Monica Blvd), Santa Monica, (90404), USA
| | - Henrik Schmidt
- Department of Clinical Medicine and Department of Oncology, Aarhus University Hospital, (Nørrebrogade 44), Aarhus C, (DK-8000), Denmark
| | - Aviram Nissan
- Department of Surgery, Hadassah Hebrew University Medical Center, (P.O.Box 24035), Jerusalem, (il-91240), Israel
| | - Laura Ridolfi
- Immunotherapy Unit, IRST Cancer Institute, (Via P. Maroncelli 40) Meldola, (47014), Italy
| | - Steinar Aamdal
- Department of Oncology, Section for Clinical Cancer Research, Oslo University Hospital, (Ullernchausseen 70, OUS Radiumhospitalet), Oslo, (0310), Norway
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, (SE-171), Stockholm, (77), Sweden
| | - Michele Guida
- Department of Medical Oncology, National Institute of Cancer, (Viale Orazio Flacco 65), Bari, (70124), Italy
| | - David M Hyams
- Desert Surgical Oncology, (39000 Bob Hope Drive), Rancho Mirage, (92270), USA
| | - Henry Gómez
- Department of Medicine, Instituto Nacional de Enfermedades Neoplásicas, (Avenida Angamos, Este 2520), Lima (34), Perú
| | - Lars Bastholt
- EORTC Melanoma Group, Odense University Hospital, (Sdr. Boulevard 29), Odense, (DK-5000), Denmark
| | - Scott D Chasalow
- Exploratory Development, Global Biometric Sciences, Bristol-Myers Squibb Company, (Route 206 & Province Line Rd), Princeton, (08543), USA
| | - David Berman
- Research and Development, Discovery Medicine, Bristol-Myers Squibb Company, (Route 206 & Province Line Rd), Princeton, (08543), USA
| |
Collapse
|
755
|
Kim JC, Yu JH, Cho YK, Jung CS, Ahn SH, Gong G, Kim YS, Cho DH. Expression of SPRR3 is associated with tumor cell proliferation in less advanced stages of breast cancer. Breast Cancer Res Treat 2011; 133:909-16. [PMID: 22076481 DOI: 10.1007/s10549-011-1868-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/31/2011] [Indexed: 12/18/2022]
Abstract
Small proline rich repeat protein 3 (SPRR3), a member of the SPRR family of cornified envelope precursor proteins, is a marker for terminal squamous cell differentiation. Previously, this laboratory showed that SPRR3 is strongly upregulated in colorectal tumors, and is involved in the tumorigenesis. The current study was performed to investigate the expression status and effect of SPRR3 in breast cancers (BCs). SPRR3 expression was examined by immunohistochemistry in 241 tumor samples from BC patients. SPRR3 was overexpressed in more than half of all BC samples. SPRR3 overexpression was significantly associated with less advanced stage (0-1 vs. II-III) and the absence of lymph node metastasis (P = 0.004 and 0.013, respectively). HER2/neu overexpression was closely correlated with SPRR3 overexpression in a multivariate analysis (OR, 3.23, P = 0.017). To assess the influence of SPRR3 on cell proliferation and related signaling pathways, SPRR3-transfected clones from the SPRR3-negative T-47D human BC cell line were generated. Among the total of six SPRR3-overexpressing clones, five showed marked proliferation compared with SPRR3-nonexpressing control cells from day 3 of culture (P < 0.001). The SPRR3-overexpressing BC clones showed increased phosphorylation of AKT and MDM2, p21 overexpression, and p53 downregulation. Furthermore, phosphorylation of MEK and MAPK was markedly increased. This study demonstrates that SPRR3 promotes BC cell proliferation by enhancing p53 degradation via the AKT and MAPK pathways and is, therefore, a potential novel therapeutic target for less advanced stages of BC.
Collapse
Affiliation(s)
- Jin Cheon Kim
- Department of Surgery, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
756
|
Abstract
This chapter presents past and present FISH techniques and specific applications of FISH. Although array technology has revolutionized cytogenetics, FISH remains indispensible. While array technology provides a high resolution screen of the entire genome for gains and losses, it does not allow for visualization of the genomic structure of gains. Thus, FISH continues to be useful as an adjunct to arrays. FISH also continues to be widely used in conjunction with banded chromosome analysis, and as a stand-alone technique for the detection of genomic alterations in neoplastic disorders.
Collapse
|
757
|
Yousif NG, Al-Amran FG. Novel Toll-like receptor-4 deficiency attenuates trastuzumab (Herceptin) induced cardiac injury in mice. BMC Cardiovasc Disord 2011; 11:62. [PMID: 21999911 PMCID: PMC3209438 DOI: 10.1186/1471-2261-11-62] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 10/14/2011] [Indexed: 12/24/2022] Open
Abstract
Background Cardiac inflammation and generation of oxidative stress are known to contribute to trastuzumab (herceptin) induced cardiac toxicity. Toll-like receptors (TLRs) are a part of the innate immune system and are involved in cardiac stress reactions. Since TLR4 might play a relevant role in cardiac inflammatory signaling, we investigated whether or not TLR4 is involved in trastuzumab induced cardiotoxicity. Methods Seven days after a single injection of herceptin (2 mg/kg; i.p.), left ventricular pressure volume loops were measured in HeN compotent (TLR4+/+) and HeJ mutant (TLR4-/-) treated with trastuzumab and control mice. Immunofluorescent staining for monocyte infiltration and analyses of plasma by (ELISAs) for different chemokines including: MCP-1and tumor necrosis factor-α (TNF-α), Western immunoblotting assay for ICAM-1, and used troponin I for cardiac injury marker. Results Trastuzumab injection resulted in an impairment of left ventricular function in TLR-4 competent (HeN), in contrast TLR4-/- trastuzumab mice showed improved left ventricular function EF%, CO; p < 0.05, attenuation of mononuclear cell infiltration in TLR4 -/-; p < 0.05 vs.TLR-4 competent (HeN), reduced level of cytokines TNF-α, MCP-1 and ICAM-1 expression in TLR4-/-, marked reduction of myocardial troponin-I levels in TLR4-deficient mice. Data are presented as means ± SE; n = 8 in each group p < 0.05 vs.TLR-4 competent (HeN). Conclusions Treatment with trastuzumab induces an inflammatory response that contributes to myocardial tissue TLR4 mediates chemokine expression (TNF-α, MCP-1and ICAM-1), so in experimental animals TLR4 deficiency improves left ventricular function and attenuates pathophysiological key mechanisms in trastuzumab induced cardiomyopathy.
Collapse
Affiliation(s)
- Nasser Ghaly Yousif
- University of Colorado Denver, Department of Medicine and Surgery, Aurora, CO 80045, USA.
| | | |
Collapse
|
758
|
Liikanen I, Monsurrò V, Ahtiainen L, Raki M, Hakkarainen T, Diaconu I, Escutenaire S, Hemminki O, Dias JD, Cerullo V, Kanerva A, Pesonen S, Marzioni D, Colombatti M, Hemminki A. Induction of interferon pathways mediates in vivo resistance to oncolytic adenovirus. Mol Ther 2011; 19:1858-66. [PMID: 21792178 PMCID: PMC3188743 DOI: 10.1038/mt.2011.144] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Oncolytic adenoviruses are an emerging experimental approach for treatment of tumors refractory to available modalities. Although preclinical results have been promising, and clinical safety has been excellent, it is also apparent that tumors can become virus resistant. The resistance mechanisms acquired by advanced tumors against conventional therapies are increasingly well understood, which has allowed development of countermeasures. To study this in the context of oncolytic adenovirus, we developed two in vivo models of acquired resistance, where initially sensitive tumors eventually gain resistance and relapse. These models were used to investigate the phenomenon on RNA and protein levels using two types of analysis of microarray data, quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry. Interferon (IFN) signaling pathways were found upregulated and Myxovirus resistance protein A (MxA) expression was identified as a marker correlating with resistance, while transplantation experiments suggested a role for tumor stroma in maintaining resistance. Furthermore, pathway analysis suggested potential therapeutic targets in oncolytic adenovirus-resistant cells. Improved understanding of the antiviral phenotype causing tumor recurrence is of key importance in order to improve treatment of advanced tumors with oncolytic adenoviruses. Given the similarities between mechanisms of action, this finding might be relevant for other oncolytic viruses as well.
Collapse
Affiliation(s)
- Ilkka Liikanen
- Cancer Gene Therapy Group, Molecular Cancer Biology Program, Transplantation Laboratory, Haartman Institute and Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
759
|
Ahn ER, Vogel CL. Dual HER2-targeted approaches in HER2-positive breast cancer. Breast Cancer Res Treat 2011; 131:371-83. [PMID: 21956210 DOI: 10.1007/s10549-011-1781-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/13/2011] [Indexed: 01/24/2023]
Abstract
Approximately 15-20% of all breast cancers are human epidermal growth factor receptor 2 (HER2) positive, with clinical studies having validated the HER2 receptor tyrosine kinase pathway as an important therapeutic target. Presently, two HER2-targeted therapies are approved by the Food and Drug Administration for treatment of HER2-positive breast cancer: the HER2-targeted humanized monoclonal antibody trastuzumab and the small-molecule tyrosine kinase inhibitor lapatinib. Despite use of these HER2-targeted agents, many patients still experience disease progression. For this reason, numerous new agents and therapeutic strategies are under investigation. Based on preclinical data suggesting synergistic effects from dual therapy targeting HER2, clinical trials that test the effects of combining anti-HER2 agents have been conducted and are ongoing. Here, we review recently presented data from several clinical trials, which indicate that the strategy of combining HER2 blockade therapies can offer greater clinical efficacy, with adverse effects of varying degrees. Specifically, we review new data reported at the 2010 San Antonio Breast Cancer Symposium (SABCS 2010), including the phase II NeoSphere and phase III NeoALTTO clinical trials, and data from three clinical trials reported at the 2011 American Society of Clinical Oncology (ASCO 2011) meeting. Together these trials elucidate the potential role of combining trastuzumab with lapatinib or pertuzumab. We also discuss additional ongoing studies that will help further define the role of dual HER2 blockade therapies and its impact on clinical practice.
Collapse
Affiliation(s)
- Eugene R Ahn
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Suite 3300, Miami, FL 33136, USA.
| | | |
Collapse
|
760
|
Ross JS. Update on HER2 testing for breast and upper gastrointestinal tract cancers. Biomark Med 2011; 5:307-18. [PMID: 21657840 DOI: 10.2217/bmm.11.31] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
With the regulatory approvals in Europe and the USA of trastuzumab-based anti-HER2 targeted therapy for upper gastrointestinal cancers in 2010, HER2 testing has now become universal for newly diagnosed cases of both breast cancer and adenocarcinomas of esophagus, stomach and gastroesophageal origin. In the 12 years or more since the approval of trastuzumab for breast cancer, general refinements in approaches to HER2 testing, including a greater understanding of the implications of preanalytic factors impacting the test results and the application of standardization of reporting of HER2 test results, have taken place. There has also been continuing development in breast cancer with the introduction of new HER2 tests, including non-FISH tests, dimerization assays, phosphorylated HER2 receptor tests, mRNA-based tests, HER2 gene sequencing tests and the application of HER2 testing to circulating tumor cells. Most recently, the introduction of HER2 testing for upper gastrointentinal malignancies has emphasized the need for performing and interpreting slide-based assays in a manner unique to these specimens and not to apply the breast cancer testing protocols to esophageal and gastric adenocarcinomas.
Collapse
|
761
|
Atkinson R, Mollerup J, Laenkholm AV, Verardo M, Hawes D, Commins D, Engvad B, Correa A, Ehlers CC, Nielsen KV. Effects of the change in cutoff values for human epidermal growth factor receptor 2 status by immunohistochemistry and fluorescence in situ hybridization: a study comparing conventional brightfield microscopy, image analysis-assisted microscopy, and interobserver variation. Arch Pathol Lab Med 2011; 135:1010-6. [PMID: 21809992 DOI: 10.5858/2010-0462-oar] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CONTEXT New guidelines for HER2 testing have been introduced. OBJECTIVES To evaluate the difference in HER2 assessment after introduction of new cutoff levels for both immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) and to compare interobserver agreement and time to score between image analysis and conventional microscopy. DESIGN Samples from 150 patients with breast cancer were scored by 7 pathologists using conventional microscopy, with a cutoff of both 10% and 30% IHC-stained cells, and using automated microscopy with image analysis. The IHC results were compared individually and to HER2 status as determined by FISH, using both the approved cutoff of 2.0 and the recently introduced cutoff of 2.2. RESULTS High concordance was found in IHC scoring among the 7 pathologists. The 30% cutoff led to slightly fewer positive IHC observations. Introduction of a FISH equivocal zone affected 4% of the FISH scores. If cutoff for FISH is kept at 2.0, no difference in patient selection is found between the 10% and the 30% IHC cutoff. Among the 150 breast cancer samples, the new 30% IHC and 2.2 FISH cutoff levels resulted in one case without a firm diagnosis because both IHC and FISH were equivocal. Automated microscopy and image analysis-assisted IHC led to significantly better interobserver agreement among the 7 pathologists, with an increase in mean scoring time of only about 30 seconds per slide. CONCLUSIONS The change in cutoff levels led to a higher concordance between IHC and FISH, but fewer samples were classified as HER2 positive.
Collapse
Affiliation(s)
- Roscoe Atkinson
- Department of Pathology, University of Southern California, Los Angeles, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
762
|
Ravi A, Bang H, Karsif K, Nori D. Breast cancer in men: prognostic factors, treatment patterns, and outcome. Am J Mens Health 2011; 6:51-8. [PMID: 21831929 DOI: 10.1177/1557988311416495] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
PURPOSE The aim of this study was to review the clinical presentation and to evaluate prognostic factors, treatment modalities, outcome, and second malignancy in male breast cancer patients. A chart review was conducted of all men treated for breast cancer between January 1991 and December 2007. Cox proportional hazards regression model and Kaplan-Meier curve were used to determine prognostic factors and plot survival probabilities. Invasive carcinoma was diagnosed in 22 patients and ductal carcinoma in situ in 7 patients. With mortality as the endpoint, tumor size indicated hazard ratio (HR) of 1.5 for each 1-cm increase in tumor size (p = .03). Overall stage and increased age were associated with increased risk of mortality (HR = 2.1, p = .055; HR = 1.09 for a 1-year increase in age, p = .08, respectively). Adjuvant radiation therapy yielded an HR of 0.1 (p = .058), indicating a favorable association with the survival. Advanced age, higher stage, and increasing tumor size were unfavorable to survival in male breast carcinoma. The benefit of adjuvant radiation therapy should be addressed in future collaborative studies.
Collapse
|
763
|
Hicks DG, Schiffhauer L. Standardized Assessment of the HER2 Status in Breast Cancer by Immunohistochemistry. Lab Med 2011. [DOI: 10.1309/lmgzz58cts0dbgtw] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
764
|
Brunicardi FC, Gibbs RA, Wheeler DA, Nemunaitis J, Fisher W, Goss J, Chen C. Overview of the development of personalized genomic medicine and surgery. World J Surg 2011; 35:1693-9. [PMID: 21424870 PMCID: PMC3281749 DOI: 10.1007/s00268-011-1056-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Personalized genomic medicine and surgery (PGMS) represents a new approach to health care that customizes patients' medical treatment according to their own genetic information. This new approach is the result of increased knowledge of the human genome and ways this information can be applied by physicians in the medical and surgical management of their patients. A patient's genotype can yield important information concerning disease susceptibility and the effectiveness of medications, therefore guiding specific, targeted imaging and treatment therapies. This review summarizes major achievements of human genomic studies and applications of genomics in health care. Five years ago we developed a model for the development of PGMS in which genomic profile guides choice of therapy. In this article we discussed our progress, including an updating of the model, and a future vision of PGMS.
Collapse
Affiliation(s)
- F Charles Brunicardi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1709 Dryden Street, Suite 1500, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
765
|
Al-azawi D, Leong S, Wong L, Kay E, Hill ADK, Young L. HER-2 positive and p53 negative breast cancers are associated with poor prognosis. Cancer Invest 2011; 29:365-9. [PMID: 21599513 DOI: 10.3109/07357907.2011.584586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
p53 and HER-2 coexpression in breast cancer has been controversial. These markers were tested using immunohistochemistry and HercepTest. HER-2 expression is related to reduced breast cancer survival (p = .02) . p53 expression relates to HER-2 expression (p = .029). Coexpression between p53 and HER-2 has no relation to prognosis. On univariate and multivariate analysis, combination of HER-2 positive and p53 negative expression was associated with a poor prognosis (p = .018 and p = .027, respectively), while the combination of HER-2 negative and p53 positive expression was associated with a favorable prognosis (p = .022 and p = .010, respectively). Therefore the expression of these markers should be considered collectively.
Collapse
|
766
|
Maresch J, Schoppmann SF, Thallinger CMR, Zielinski CC, Hejna M. Her-2/neu gene amplification and over-expression in stomach and esophageal adenocarcinoma: from pathology to treatment. Crit Rev Oncol Hematol 2011; 82:310-22. [PMID: 21783379 DOI: 10.1016/j.critrevonc.2011.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 06/24/2011] [Indexed: 12/28/2022] Open
Abstract
Discovery of the over-expression of Her-2/neu or the amplification of its regulatory gene in stomach and esophageal cancer has resulted in targeted treatment directed at this protein. The fact itself and its consequences have been the topic of an abundance of studies and clinical trials. In the present report we review the current state of the art as regards diagnosis of the over-expression and amplification of Her-2/neu, its inhibition as a new therapeutic concept, treatment toxicity, and the development of resistance to Her-2/neu as a limiting factor in stomach and esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Judith Maresch
- Department of Pathology, General Hospital - Medical University of Vienna, Austria
| | | | | | | | | |
Collapse
|
767
|
Valachis A, Mauri D, Polyzos NP, Chlouverakis G, Mavroudis D, Georgoulias V. Trastuzumab combined to neoadjuvant chemotherapy in patients with HER2-positive breast cancer: a systematic review and meta-analysis. Breast 2011; 20:485-90. [PMID: 21784637 DOI: 10.1016/j.breast.2011.06.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 04/19/2011] [Accepted: 06/29/2011] [Indexed: 10/18/2022] Open
Abstract
PURPOSE To perform a meta-analysis in order to quantify the actual cumulative randomized evidence for the benefit and toxicity of trastuzumab combined with neoadjuvant chemotherapy in HER2-positive breast cancer. METHODS Potentially eligible trials were located through PubMed and Cochrane Library searches and abstracts of major international conferences. The endpoints that we assessed were pathologic complete response (pCR) rate, breast-conserving surgery (BCS) rate and toxicity. RESULTS Five trials were identified with 515 eligible patients. The probability to achieve pCR was higher for the trastuzumab plus chemotherapy arm (RR 1.85, 95% CI: 1.39-2.46; p-value < 0.001). No significant difference in terms of breast-conserving surgery between the two treatment arms was observed (OR: 0.98, 95% CI: 0.80-1.19, p-value = 0.82). Regarding toxicity, the addition of trastuzumab did not increase the incidence of neutropenia, neutropenic fever, and cardiac adverse events. CONCLUSION The addition of trastuzumab in HER2-positive breast cancer in the neoadjuvant setting improves the probability of achieving higher pCR with no additional toxicity. Based on the available evidence, the use of trastuzumab combined with neoadjuvant chemothetherapy in patients with HER2-positive breast cancer seems to offer substantial benefit in terms of pCR.
Collapse
Affiliation(s)
- Antonis Valachis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece.
| | | | | | | | | | | |
Collapse
|
768
|
Selective formation of covalent protein heterodimers with an unnatural amino acid. ACTA ACUST UNITED AC 2011; 18:299-303. [PMID: 21439474 DOI: 10.1016/j.chembiol.2011.01.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 01/07/2011] [Accepted: 01/11/2011] [Indexed: 10/18/2022]
Abstract
We report a strategy for the generation of heterodimeric protein conjugates using an unnatural amino acid with orthogonal reactivity. This paper addresses the challenges of site-specificity and homogeneity with respect to the synthesis of bivalent proteins and antibody-drug conjugates. There are numerous antibody-drug conjugates in preclinical and clinical development, yet these are based either on nonspecific lysine coupling chemistry or on disulfide modification made difficult by the large number of cysteines in antibodies. Here, we describe a recombinant approach that can be used to rapidly generate a variety of constructs with defined conjugation sites. Moreover, this methodology results in homogeneous antibody conjugates whose biological, physical, and pharmacological properties can be quantitatively assessed and subsequently optimized. As proof of concept, we have generated anti-Her2 Fab-Saporin conjugates that demonstrate excellent potency in vitro.
Collapse
|
769
|
Ross JS, Mulcahy M. HER2 Testing in Gastric/Gastroesophageal Junction Adenocarcinomas: Unique Features of a Familiar Test. GASTROINTESTINAL CANCER RESEARCH : GCR 2011; 4:106-8. [PMID: 21673877 DOI: 10.1152/ajpcell.00650.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 02/02/2011] [Indexed: 11/22/2022]
Abstract
Using the standard slide-based techniques of immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), it has been firmly established that human epidermal growth factor receptor 2 (HER2) is overexpressed in adenocarcinoma of the upper gastrointestinal tract. In the ToGA trial, the addition of the monoclonal antibody trastuzumab to a standard regimen of cisplatin and fluoropyrimidine resulted in a clinically and statistically significant benefit in terms of response rate, median progression-free survival, and median overall survival in HER2-positive patients. Major differences exist, however, between HER2 testing in gastric/gastroesophageal junction (GEJ) cancer versus breast cancer, and the ToGA trial employed a significant modification of the breast cancer scoring criteria. As trastuzumab approaches regulatory approval in the United States for gastric/GEJ cancer, it is critical that pathologists and diagnostic laboratories learn and apply the unique criteria for assessing gastric/GEJ tumors for their HER2 status defined by the ToGA investigators, as they ready themselves for the approximately 50,000 new specimens that will be tested for HER2 status by both IHC and FISH each year.
Collapse
|
770
|
Quantum dots-based molecular classification of breast cancer by quantitative spectroanalysis of hormone receptors and HER2. Biomaterials 2011; 32:7592-9. [PMID: 21745686 DOI: 10.1016/j.biomaterials.2011.06.029] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 06/10/2011] [Indexed: 02/07/2023]
Abstract
The emerging molecular breast cancer (BC) classification based on key molecules, including hormone receptors (HRs), and human epidermal growth factor receptor 2 (HER2) has been playing an important part of clinical practice guideline. The current molecular classification mainly based on their fingerprints, however, could not provide enough essential information for treatment decision making. The molecular information on both patterns and quantities could be more helpful to heterogeneities understanding for BC personalized medicine. Here we conduct quantitative determination of HRs and HER2 by quantum dots (QDs)-based quantitative spectral analysis, which had excellent consistence with traditional method. Moreover, we establish a new molecular classification system of BC by integrating the quantitative information of HER2 and HRs, which could better reveal BC heterogeneity and identify 5 molecular subtypes with different 5-year prognosis. Furthermore, the emerging 5 molecular subtypes based on simple quantitative molecules information could be as informative as multi-genes analysis in routine practice, and might help formulate a more personalized comprehensive therapy strategy and prognosis prediction.
Collapse
|
771
|
Jørgensen JT, Møller S, Rasmussen BB, Winther H, Schønau A, Knoop A. High concordance between two companion diagnostics tests: a concordance study between the HercepTest and the HER2 FISH pharmDx kit. Am J Clin Pathol 2011; 136:145-151. [PMID: 21685042 DOI: 10.1309/ajcpjpj8zwgdttwc] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The present study was done to investigate the concordance between the HER2 status measured by immunohistochemical analysis (HercepTest, DAKO, Carpinteria, CA) and fluorescence in situ hybridization (FISH; HER2 FISH pharmDx, DAKO) in a large study cohort (n = 681) of patients with high-risk breast cancer. A high agreement between immunohistochemical and FISH results was demonstrated. For the whole study population, the agreement between the 2 assays was 93.1% with a corresponding κ coefficient of 0.85. When the equivocal immunohistochemical 2+ cases were excluded from the analysis (n = 79), the agreement increased to 95.0% with a κ coefficient of 0.90. When the cutoff value for amplified/nonamplified cases in the HER2 FISH assay was increased from 2.0 to 2.2 as recommended in the American Society of Clinical Oncology/College of American Pathologists guidelines, the concordance between the 2 assays was 94.3% with a κ coefficient of 0.87 in the whole study population. When the equivocal immunohistochemical 2+ cases were excluded from this analysis, the concordance is similar (95.7% with a κ coefficient of 0.91).
Collapse
|
772
|
Gruver AM, Portier BP, Tubbs RR. Molecular pathology of breast cancer: the journey from traditional practice toward embracing the complexity of a molecular classification. Arch Pathol Lab Med 2011; 135:544-57. [PMID: 21526953 DOI: 10.5858/2010-0734-rair.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Adenocarcinoma of the breast is the most frequent cancer affecting women in both developed and developing regions of the world. From the moment of clinical presentation until the time of pathologic diagnosis, patients affected by this disease will face daunting questions related to prognosis and treatment options. While improvements in targeted therapies have led to increased patient survival, these same advances have created the imperative to accurately stratify patients to achieve maximum therapeutic efficacy while minimizing side effects. In this evolving era of personalized medicine, there is an ever-increasing need to overcome the limitations of traditional diagnostic practice. OBJECTIVE To summarize the molecular diagnostics traditionally used to guide prognostication and treatment of breast carcinomas, to highlight published data on the molecular classification of these tumors, and to showcase molecular assays that will supplement traditional methods of categorizing the disease. DATA SOURCES A review of the literature covering the molecular diagnostics of breast carcinomas with a focus on the gene expression and array studies used to characterize the molecular signatures of the disease. Special emphasis is placed on summarizing evolving technologies useful in the diagnosis and characterization of breast carcinoma. CONCLUSIONS Available and emerging molecular resources will allow pathologists to provide superior diagnostic, prognostic, and predictive information about individual breast carcinomas. These advances should translate into earlier identification and tailored therapy and should ultimately improve outcome for patients affected by this disease.
Collapse
Affiliation(s)
- Aaron M Gruver
- Department of Molecular Pathology, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
773
|
Houssami N, Macaskill P, Balleine RL, Bilous M, Pegram MD. HER2 discordance between primary breast cancer and its paired metastasis: tumor biology or test artefact? Insights through meta-analysis. Breast Cancer Res Treat 2011; 129:659-74. [PMID: 21698410 DOI: 10.1007/s10549-011-1632-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 06/03/2011] [Indexed: 12/31/2022]
Abstract
The proto-oncogene, HER2, has prognostic and predictive relevance in invasive breast cancer (IBC). HER2 testing of primary IBC guides treatment selection and is assumed to reflect HER2 status of associated metastases, although HER2 discordance between IBC and metastasis has been reported. Systematic review and meta-analysis of HER2 status in IBC and its paired loco-regional or distant metastasis were done. Quality appraisal considered whether (within-subject) testing conditions were maintained for paired primary and metastasis. Random effects logistic regression models were used to estimate pooled within-subject HER2 discordant proportions and to examine study-level covariates, including tumor-related and testing-related variables, potentially associated with HER2 discordance differences across (between) studies. Modelled paired HER2 data for primary and metastatic cancer (2520 subjects, 26 studies) showed a pooled HER2 discordance of 5.5% (3.6-8.5%). Sensitivity analysis, excluding the only study not maintaining same conditions for paired testing, gave a pooled estimate of 5.2% (3.5-7.8%). Pooled discordant proportion was not associated with differences between studies in test type, test scoring or interpretation criteria, subjects' median age, study time-frame, or HER2 positivity in primary cancer (all P > 0.05). However, type of metastasis was significantly associated with estimated HER2 discordance (P = 0.0017): studies of primary tumor paired with distant metastases had higher discordance [11.5% (6.9-18.6%)] than studies of primary paired with lymph node metastases only [4.1% (2.4-7.2%)], or those paired with nodal or various metastases [3.3% (2.0-5.6%)]; P < 0.01. HER2 discordant proportion was higher where paired metastases were metachronous relative to synchronous to primary IBC (P = 0.0024). Sensitivity analysis provided weak evidence (P = 0.074) that discordance in the direction of change from HER2-negative primary cancer to HER2-positive paired metastasis was more likely than the reverse. Study-level meta-analysis suggests factors associated with the type of metastasis as underlying mechanisms for observed HER2 discordance between primary IBC and paired metastasis. Test-related factors did not account for differences across studies in the HER2 discordant proportion.
Collapse
Affiliation(s)
- Nehmat Houssami
- Screening and Test Evaluation Program, School of Public Health (A27), Sydney Medical School, University of Sydney, Sydney 2006, Australia.
| | | | | | | | | |
Collapse
|
774
|
Zagozdzon R, Gallagher WM, Crown J. Truncated HER2: implications for HER2-targeted therapeutics. Drug Discov Today 2011; 16:810-6. [PMID: 21704182 DOI: 10.1016/j.drudis.2011.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/05/2011] [Accepted: 06/06/2011] [Indexed: 11/15/2022]
Abstract
The HER2 receptor is currently one of the flagship therapeutic targets in clinical oncology. Trastuzumab, an antibody targeting HER2, has become a foundation of care in women with HER2-positive breast cancer. However, many women with metastatic breast cancer do not respond to trastuzumab-based therapy. One possible source of trastuzumab resistance is the presence of truncated forms of HER2 in the tumor. Numerous studies suggest that detection of truncated HER2 in the tumor should result in modification of the classical therapeutic approach. Recent development of several promising compounds brings hope that a generation of novel therapeutic modalities against HER2-positive cancers will be delivered in the future.
Collapse
Affiliation(s)
- Radoslaw Zagozdzon
- Cancer Biology and Therapeutics Group, UCD Conway Institute, UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.
| | | | | |
Collapse
|
775
|
Dutt A, Ramos AH, Hammerman PS, Mermel C, Cho J, Sharifnia T, Chande A, Tanaka KE, Stransky N, Greulich H, Gray NS, Meyerson M. Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer. PLoS One 2011; 6:e20351. [PMID: 21666749 PMCID: PMC3110189 DOI: 10.1371/journal.pone.0020351] [Citation(s) in RCA: 303] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 04/30/2011] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Squamous cell lung carcinomas account for approximately 25% of new lung carcinoma cases and 40,000 deaths per year in the United States. Although there are multiple genomically targeted therapies for lung adenocarcinoma, none has yet been reported in squamous cell lung carcinoma. METHODOLOGY/PRINCIPAL FINDINGS Using SNP array analysis, we found that a region of chromosome segment 8p11-12 containing three genes-WHSC1L1, LETM2, and FGFR1-is amplified in 3% of lung adenocarcinomas and 21% of squamous cell lung carcinomas. Furthermore, we demonstrated that a non-small cell lung carcinoma cell line harboring focal amplification of FGFR1 is dependent on FGFR1 activity for cell growth, as treatment of this cell line either with FGFR1-specific shRNAs or with FGFR small molecule enzymatic inhibitors leads to cell growth inhibition. CONCLUSIONS/SIGNIFICANCE These studies show that FGFR1 amplification is common in squamous cell lung cancer, and that FGFR1 may represent a promising therapeutic target in non-small cell lung cancer.
Collapse
Affiliation(s)
- Amit Dutt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Center, Kharghar, Navi Mumbai, India
| | - Alex H. Ramos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter S. Hammerman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Craig Mermel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
| | - Jeonghee Cho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Tanaz Sharifnia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
| | - Ajit Chande
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Center, Kharghar, Navi Mumbai, India
| | - Kumiko Elisa Tanaka
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
| | - Nicolas Stransky
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
| | - Heidi Greulich
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
776
|
Brügmann A, Sorensen BS. Identifying responders to trastuzumab therapy in breast cancer. Future Oncol 2011; 7:767-73. [DOI: 10.2217/fon.11.44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In breast cancer, HER2-targeted therapy with trastuzumab has gained significant attention, owing to the dramatic response observed in a subset of HER2-positive patients. The mechanisms of action are complex and not fully understood, and much effort has been spent in order to identify responders. Good patient management, side effects of the humanized monoclonal antibody and socioeconomics all demand that the drug should be administered only to the patients who will benefit from it. This has been a difficult task and contributions to solve it have been proposed from a variety of research. In this article we describe some of these contributions based on the literature and provide our viewpoint as to which identifiers will emerge in the following decade.
Collapse
Affiliation(s)
- Anja Brügmann
- Institute of Pathology, Aalborg Hospital, Ladegaardsgade 3, 9000 Aalborg, Denmark
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Norrebrogade 44, DK 8000 Aarhus C, Denmark
| |
Collapse
|
777
|
|
778
|
Witteles RM, Fowler MB, Telli ML. Chemotherapy-associated cardiotoxicity: how often does it really occur and how can it be prevented? Heart Fail Clin 2011; 7:333-44. [PMID: 21749885 DOI: 10.1016/j.hfc.2011.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cardiotoxicity remains the limiting factor for many forms of cancer therapy and is the focus of growing research and clinical emphasis. This article outlines the current clinical evidence for left ventricular dysfunction and heart failure for the two most important classes of cardiotoxic chemotherapeutic agents, examines the potential pitfalls that have led to underestimated rates of left ventricular dysfunction from these agents, and reviews strategies for screening for and providing prophylaxis against chemotherapy-associated left ventricular dysfunction.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Biomarkers, Pharmacological
- Cardiac Imaging Techniques/methods
- Cardiotoxins
- Clinical Trials as Topic
- Drug Monitoring/methods
- Humans
- Lapatinib
- Neoplasms/drug therapy
- Outcome and Process Assessment, Health Care
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/adverse effects
- Quinazolines/administration & dosage
- Quinazolines/adverse effects
- Severity of Illness Index
- Trastuzumab
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/diagnosis
- Ventricular Dysfunction, Left/epidemiology
- Ventricular Dysfunction, Left/prevention & control
Collapse
Affiliation(s)
- Ronald M Witteles
- Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
779
|
Trastuzumab has anti-metastatic and anti-angiogenic activity in a spontaneous metastasis xenograft model of esophageal adenocarcinoma. Cancer Lett 2011; 308:54-61. [PMID: 21570176 DOI: 10.1016/j.canlet.2011.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/15/2011] [Accepted: 04/18/2011] [Indexed: 01/02/2023]
Abstract
HER-2/neu over-expression occurs in 10-40% of patients with esophageal adenocarcinoma. Therefore, inhibitory effects of trastuzumab on proliferation, neoangiogenesis and metastatic spread of the esophageal adenocarcinoma cell line PT1590 were investigated (subcutaneous xenograft model). PT1590 revealed an amplified copy number of c-erbB2 and HER-2/neu over-expression occured in xenograft tumors and spontaneous lung metastases. PT1590 proliferation was significantly inhibited by trastuzumab in vitro. In vivo, tumor weight, volume, microvessel density and number of lung metastases decreased significantly after three weeks of treatment. These data suggest the importance of HER-2/neu for metastatic spread in esophageal adenocarcinoma and encourages clinical trials.
Collapse
|
780
|
Ross JS. Point: Fluorescence in situ hybridization is the preferred approach over immunohistochemistry for determining HER2 status. Clin Chem 2011; 57:980-2. [PMID: 21558455 DOI: 10.1373/clinchem.2010.160762] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jeffrey S Ross
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
781
|
Penault-Llorca F, Chenard MP, Bouché O, Emile JF, Bibeau F, Metges JP, André T, Monges G. [HER2 and gastric cancer. Recommendations for clinical practice in 2011]. Ann Pathol 2011; 31:78-87. [PMID: 21601111 DOI: 10.1016/j.annpat.2011.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 03/09/2011] [Accepted: 03/12/2011] [Indexed: 10/18/2022]
Abstract
Trastuzumab in combination with capecitabine or 5-fluorouracil and cisplatin has been approved by the European Medicines Agency (EMEA) for the treatment of patients with human epidermal growth factor receptor 2 (HER2)-positive (immunohistochemistry [IHC] 3+ or IHC 2+/ fluorescence in situ hybridization [FISH]-positive or IHC 2+/ silver in situ hybridization [SISH]-positive) metastatic adenocarcinoma of the stomach or gastro-esophageal (GE) junction. HER2 testing in gastric cancer (GC) differs from testing in breast cancer (BC) due to major differences in the tumor biology; as the disease is progressing rapidely, we recommend to test every GC at diagnosis and to offer a rapid testing (less than five days) in the metastatic setting. IHC should be the initial testing methodology and FISH or SISH should be used to retest IHC 2+ samples. As GC more frequently shows incomplete membrane staining and focal staining for HER2, HER2 testing guidelines have been adapted from BC protocols. The scoring system is slightly different in respect to the characteristics of GC. For in situ hybridization, SISH should be used in order to identify heterogeneous staining with a higher accuracy than FISH. Enrollment in training and quality assurance programs is highly recommended. In case of negativity on biopsy, it is recommended to retest for HER2, when possible, on surgical specimens and/or metastasis. This will ensure accurate and consistent HER2 testing results, which will allow the appropriate selection of patients eligible for treatment with trastuzumab.
Collapse
Affiliation(s)
- Frédérique Penault-Llorca
- Département de pathologie, centre Jean-Perrin, BP 392, 58, rue Montalembert, 63011 Clermont-Ferrand cedex, France; EA 4233, université d'Auvergne, 63000 Clermont-Ferrand cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
782
|
Tzimagiorgis G, Michailidou EZ, Kritis A, Markopoulos AK, Kouidou S. Recovering circulating extracellular or cell-free RNA from bodily fluids. Cancer Epidemiol 2011; 35:580-9. [PMID: 21514265 DOI: 10.1016/j.canep.2011.02.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 02/28/2011] [Accepted: 02/28/2011] [Indexed: 12/18/2022]
Abstract
The presence of extracellular circulating or cell-free RNA in biological fluids is becoming a promising diagnostic tool for non invasive and cost effective cancer detection. Extracellular RNA or miRNA as biological marker could be used either for the early detection and diagnosis of the disease or as a marker of recurrence patterns and surveillance. In this review article, we refer to the origin of the circulating extracellular RNA, we summarise the data on the biological fluids (serum/plasma, saliva, urine, cerebrospinal fluid and bronchial lavage fluid) of patients suffering from various types of malignancies reported to contain a substantial amount of circulating extracellular (or cell-free) RNAs and we discuss the appropriate reagents and methodologies needed to be employed in order to obtain RNA material of high quality and integrity for the majority of the experimental methods used in RNA expression analysis. Furthermore, we discuss the advantages and disadvantages of the RT-PCR or microarray methodology which are the methods more often employed in procedures of extracellular RNA analysis.
Collapse
Affiliation(s)
- Georgios Tzimagiorgis
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 540 06 Thessaloniki, Greece.
| | | | | | | | | |
Collapse
|
783
|
Hamberg P, Bos MMEM, Braun HJJ, Stouthard JML, van Deijk GA, Erdkamp FLG, van der Stelt-Frissen IN, Bontenbal M, Creemers GJM, Portielje JEA, Pruijt JFM, Loosveld OJL, Smit WM, Muller EW, Schmitz PIM, Seynaeve C, Klijn JGM. Randomized phase II study comparing efficacy and safety of combination-therapy trastuzumab and docetaxel vs. sequential therapy of trastuzumab followed by docetaxel alone at progression as first-line chemotherapy in patients with HER2+ metastatic breast cancer: HERTAX trial. Clin Breast Cancer 2011; 11:103-13. [PMID: 21569996 DOI: 10.1016/j.clbc.2011.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 07/31/2010] [Accepted: 10/10/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND Because chemotherapy for metastatic breast cancer (MBC) is associated with relevant toxicity, sequential monotherapy trastuzumab followed by cytotoxic therapy at disease progression might be an attractive approach. METHODS In a multicenter phase II trial, 101 patients with overexpression of human epidermal growth factor receptor 2 (HER2(+)) MBC were randomized between combination-therapy trastuzumab (Herceptin) plus docetaxel (H+D) and sequential therapy of single-agent trastuzumab followed at disease progression by docetaxel alone (H→D) as first-line chemotherapy for metastatic disease. The primary endpoint was progression-free survival (PFS) after completed sequential or combination therapy. RESULTS For the H+D group the median PFS was 9.4 vs. 9.9 months for the H→D group and 1-year PFS rates were 44% vs. 35%, respectively. However the overall response rates (ORRs) were 79% vs. 53%, respectively (P = .016), and overall survival was 30.5 vs. 19.7 months, respectively (P = .11). In the H→D group, response rates to monotherapy trastuzumab and subsequent docetaxel were 34% and 39%, respectively, with a median PFS during single-agent trastuzumab of 3.9 months. The incidence and severity of neuropathy were significantly higher in the H+D group. Retrospective analysis of trastuzumab treatment beyond progression (applied in 46% of patients in the H+D group and 37% in the H→D group) showed a correlation with longer overall survival in both treatment arms (36.0 vs. 18.0 months and 30.3 vs. 18.6 months, respectively). CONCLUSION First-line treatment in patients with MBC with H→D resulted in a similar PFS compared with H+D, but the response rate was lower and the overall survival nonsignificantly shorter.
Collapse
Affiliation(s)
- Paul Hamberg
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
784
|
Thang VH, Tani E, Van TT, Krawiec K, Skoog L. HER2 status in operable breast cancers from Vietnamese women: Analysis by immunohistochemistry (IHC) and automated silver enhanced in situ hybridization (SISH). Acta Oncol 2011; 50:360-6. [PMID: 21338271 DOI: 10.3109/0284186x.2010.547217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND HER2 amplification has been reported to occur in 19-30% of breast cancers in women from Western countries. Little is known about the HER2 status in breast cancers from Vietnamese women. The aim of this study was to assess the HER2 status in Vietnamese women with operable breast cancer using immunohistochemistry and silver in situ hybridization techniques. MATERIAL AND METHOD Tissue blocks from 242 Vietnamese women with invasive primary breast carcinoma were stained by immunohistochemistry (IHC) and a dual silver in situ hybridization (SISH) for assessing HER2 status. The analysis followed international recommendations with a semi-quantitative grading of the reaction in four levels, "0", "1+", "2+" and "3+". The HER2 gene amplification was assessed by calculating the ratio of HER2/chromosome 17 in 20 tumor cell nuclei. A ratio of <1.8 was classified as non-amplification and a ratio >2.2 indicated tumors with gene amplification. A ratio between 1.8 and 2.2 was equivocal. RESULTS Using IHC, 39% of the tumors strongly expressed "3+" the HER2 protein. An intermediate level "2+" of the protein was found in 11% while 50% showed no or low "0/1+" levels of protein expression. Gene amplification was found in 95% and 46% of the tumors with high "3+" and intermediate "2+" levels of protein expression, respectively. Thus, 41% of the patients had tumors with HER2 amplification. The concordance between IHC and SISH was 87%. Gene amplification was more frequent in ER negative than in ER positive tumors, 57% versus 33%, respectively. Additionally, tumors from postmenopausal women were amplified in 55% as compared to 36% in premenopausal women. CONCLUSIONS HER2 gene amplification was found in 41% of Vietnamese breast cancers; this level is considerably higher than that previously found in a series of Swedish breast cancer women. The HER2-positive tumors were more often found in post-menopausal women than in ER negative tumors.
Collapse
Affiliation(s)
- Vu Hong Thang
- Oncology Department, Hanoi Medical University, Vietnam
| | | | | | | | | |
Collapse
|
785
|
Elnashar AT, Ali ESM, Gaber A. The prognostic value of triple negative in stage II/III breast cancer. J Oncol Pharm Pract 2011; 18:68-75. [PMID: 21422149 DOI: 10.1177/1078155211398299] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Breast cancer is no longer seen as a single disease but rather a multifaceted disease composed of distinct biological subtypes with diverse natural history, clinical, pathological, and molecular features. Recent attention has been directed at the molecular classification of breast cancer. OBJECTIVE To evaluate the prognostic value of triple-negative subtype in stage II/III breast cancer and to define the role of clinical stage in prognosis of breast cancer. METHODS We used the immunohistochemical technique to divide 255 cases of breast cancer, stages II and III, into four subtypes according to estrogen receptor/progesterone receptor and Her-2 expression. RESULTS Triple-negative subtype comprised 76.5% of the cases with 12.3% recurrence rate. Luminal A subtype also carried a poor outcome with 16.7% recurrence rate. CONCLUSION Triple-negative subtype has the worst overall and disease-free survival in stage II/III breast cancer. Clinical stage is still an independent prognostic factor in the breast cancers of all types.
Collapse
|
786
|
Rana V, Swaby RF. Targeted Therapies for HER2 Breast Cancer: A View of the Landscape. CURRENT BREAST CANCER REPORTS 2011. [DOI: 10.1007/s12609-010-0037-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
787
|
Vrolix K, Niks EH, Le Panse R, van Ostaijen-ten Dam MM, Muris AH, Jol-van der Zijde CM, van Tol MJ, Losen M, Molenaar PC, van Zoelen EJ, Berrih-Aknin S, De Baets MH, Verschuuren JJ, Martínez-Martínez P. Reduced thymic expression of ErbB receptors without auto-antibodies against synaptic ErbB in myasthenia gravis. J Neuroimmunol 2011; 232:158-65. [DOI: 10.1016/j.jneuroim.2010.10.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 09/27/2010] [Accepted: 10/21/2010] [Indexed: 11/15/2022]
|
788
|
Torrisi R, Cardillo A, Cancello G, Dellapasqua S, Balduzzi A, Ghisini R, Luini A, Veronesi P, Viale G, Goldhirsch A, Colleoni M. Phase II trial of combination of pegylated liposomal doxorubicin, cisplatin, and infusional 5-fluorouracil (CCF) plus trastuzumab as preoperative treatment for locally advanced and inflammatory breast cancer. Clin Breast Cancer 2011; 10:483-8. [PMID: 21147693 DOI: 10.3816/cbc.2010.n.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Pegylated liposomal doxorubicin (PLD) was shown as active but less toxic compared to doxorubicin in advanced breast cancer. Given its low cardiotoxicity, the combination of PLD and trastuzumab appears most attractive in the treatment of human epidermal factor receptor 2 (HER2)-positive breast cancer. PATIENTS AND METHODS We investigated the activity of 8 courses of PLD in combination with cisplatin and infusional 5-fluorouracil (CCF) plus 3-week trastuzumab in patients with primary or recurrent cT2-T4 a-d, N0-3, M0 any estrogen receptor (ER), HER2-positive breast cancer. Patients with ER and/or progesterone receptor (PgR) ≥ 10% tumors received also letrozole (plus triptorelin if premenopausal). The principal endpoint was clinical response rate; secondary endpoints were the pathologic complete response rate (pCR) and the cardiac safety of the combination. RESULTS Thirty-two patients were enrolled in the study and all are evaluable for response and toxicity. Fifteen patients (47%) had ER-positive tumors, 15 patients and 2 patients had ER absent and ER poor tumors, respectively. Thirteen patients (41%) had inflammatory breast cancer (IBC) and 84% of patients had clinically positive nodes. A clinical response rate of 94% (95% CI, 79%-99%) and a pCR rate of 41% (95% CI, 24%-59%) were observed. Fifty-four percent of patients with IBC obtained a pCR. Eleven patients discontinued treatment before completing 8 courses as planned. No patient developed relevant cardiac toxicity. CONCLUSION In this series of very locally advanced breast cancer, the combination of CCF and trastuzumab was very active obtaining an impressive rate of pCR, particularly in IBC, which merits further investigation in larger series.
Collapse
Affiliation(s)
- Rosalba Torrisi
- Research Unit of Medical Senology, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
789
|
Jha K, Shukla M, Pandey M. Survivin expression and targeting in breast cancer. Surg Oncol 2011; 21:125-31. [PMID: 21334875 DOI: 10.1016/j.suronc.2011.01.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/27/2010] [Accepted: 01/08/2011] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Survivin a multifunctional protein that controls cell division, inhibition of apoptosis and promotion of angiogenesis. It is expressed in most human neoplasm, but is absent in normal and differentiated tissues. The purpose of this article is to overview the expression of survivin, effect of its expression in response to treatment, correlation with other markers and newer advancement in targeting survivin. METHODS A detailed search of Medline was carried out using the following search strategy: "((survivin) OR ((apoptosis) AND (inhibitor OR inhibitors))) AND ((breast) AND (neoplasm OR neoplasms OR tumor OR tumor OR cancer OR carcinoma))". Abstract of all articles thus identified were reviewed to identify the relevant studies, full articles of studies thus identified were then obtained and reviewed. All relevant data was extracted and tabulated. RESULTS Survivin expression by Immunohistochemistry was identified in 65.3% (55.2-90.0%) of the breast cancer patients among the identified studies while survivin mRNA by RT-PCR was identified in 93.6% (90-97%). Survivin expression has been reported to be associated with over expression of HER 2, vascular endothelial growth factor (VEGF), urokinase plasminogen activator (uPA)/PAI-1. CONCLUSION Survivin is over expressed in majority of breast cancers. The over expression of survivin is found to correlate with HER 2 and EGFR expression. Survivin expression has been found to confer resistance to chemotherapy and radiation. Targeting survivin in experimental models improves survival. More studies are needed on the role of survivin in multi drug resistance (MDR) in the presence of Pgp/uPA/PAI-1 and the impact of survivin over expression in triple negative breast cancer.
Collapse
Affiliation(s)
- Kumkum Jha
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | | |
Collapse
|
790
|
Soria JC, Blay JY, Spano JP, Pivot X, Coscas Y, Khayat D. Added value of molecular targeted agents in oncology. Ann Oncol 2011; 22:1703-16. [PMID: 21300696 DOI: 10.1093/annonc/mdq675] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The treatment of certain cancers has been revolutionised in recent years by the introduction of novel drugs designed to target specific molecular factors implicated in tumour growth. Notable examples include trastuzumab, a humanized monoclonal antibody (mAb) against human epidermal growth factor receptor (HER)-2 in women with HER2-positive breast cancer; rituximab, an anti-CD20 mAb in patients with non-Hodgkin's lymphoma; imatinib, a tyrosine kinase inhibitor in KIT-positive gastrointestinal stromal tumours and sunitinib, another tyrosine kinase inhibitor, in metastatic renal cell carcinoma. For regulatory reasons, new molecular targeted agents are first evaluated in advanced and metastatic disease, wherein they prolong survival. However, their most profound impact has been observed in the adjuvant setting, where they may contribute to curative therapy rather than mere palliation. Expansion in the use of molecular targeted therapies will have important cost implications for health care systems. Although expensive, on a monthly basis, molecular targeted therapies may not be more costly than treatments for other major chronic diseases, especially considering the contribution of cancer to the global disease burden, the associated socioeconomic costs and the long-term benefits of therapy. Nevertheless, the use of these agents must be optimised, in part using molecular biomarkers associated with drug response.
Collapse
Affiliation(s)
- J C Soria
- Institut Gustave Roussy, Villejuif, France.
| | | | | | | | | | | |
Collapse
|
791
|
Lefever M, Kosmeder JW, Farrell M, Bieniarz C. Microwave-mediated synthesis of labeled nucleotides with utility in the synthesis of DNA probes. Bioconjug Chem 2011; 21:1773-8. [PMID: 20812713 DOI: 10.1021/bc100013b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A novel method of linking haptens to deoxycytidine 5'-triphosphate via microwave-mediated bisulfate-catalyzed transamination with hydrazine has been developed. This method enables the tethering of small molecule haptens to dCTP via a discrete polyethylene glycol (PEG) spacer, yielding N(4)-aminodeoxycytidine 5'-triphosphate-dPEG-haptens. This synthetic approach employs microwave-catalyzed hydrazinolysis that enables the attachment of spacers via hydrazine linkages. The microwave-mediated introduction of this hydrazine handle provides a significant improvement in yield over those of published thermal methods. The microwave reaction was shown to be scalable, and the final product was amenable to labeling with a wide variety of haptens. The resulting nucleotide triphosphates, N(4)-aminodeoxycytidine 5'-triphosphate-dPEG-haptens, can serve as unique substrates for the enzyme-mediated labeling of DNA probes. The efficacy of incorporation of one such novel nucleotide, N(4)-aminodeoxycytidine 5'-triphosphate-dPEG(4)-DNP, has been demonstrated in nick translation labeling of HER2 and HPV probes. The labeled probes have been shown to be effective in visualizing their target genes in tissue.
Collapse
Affiliation(s)
- Mark Lefever
- Ventana Medical Systems, Inc., Tucson, Arizona 85755, USA
| | | | | | | |
Collapse
|
792
|
Results and implications of the Trastuzumab for Gastric Cancer (ToGA) trial. ACTA ACUST UNITED AC 2011. [DOI: 10.4155/cli.10.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
793
|
Wong EHF, Fox JC, Ng MYM, Lee CM. Toward personalized medicine in the neuropsychiatric field. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 101:329-49. [PMID: 22050858 DOI: 10.1016/b978-0-12-387718-5.00013-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
There are great expectations for the personalized medicine approach to address the therapeutic needs of patients in the twenty-first century. Advances in human genome science and molecular innovations in neuroscience have encouraged the pharmaceutical industry to focus beyond broad spectrum population therapeutics--the driving force behind the "blockbuster" product concept--to personalized medicine. For central nervous system (CNS) therapeutics, repeated failures in converting scientific discoveries to clinical trial successes and regulatory approvals have precipitated a drug pipeline crisis and eroded confidence in the industry. This chapter describes how innovations in genomics and translational medicine can impact the future of neuropsychiatry and deconvolute the complexity of psychiatric diseases from symptoms biology. A targeted and consistent investment is needed to restore confidence in translating science into clinical success.
Collapse
Affiliation(s)
- Erik H F Wong
- AstraZeneca Pharmaceuticals, External Science, CNS-Pain Innovative Medicine Unit, Wilmington, Delaware, USA
| | | | | | | |
Collapse
|
794
|
Shin HC, Bae YK, Kim A, Park SJ. HER2-Positive Breast Carcinomas with Co-amplification or Gain of Chromosome 17 Centromere Locus: Report of Three Cases and an Impact on HER2 Testing. KOREAN JOURNAL OF PATHOLOGY 2011. [DOI: 10.4132/koreanjpathol.2011.45.6.665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Hyeong Chan Shin
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Aeri Kim
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Seok Ju Park
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
795
|
Shah S, Chen B. Testing for HER2 in Breast Cancer: A Continuing Evolution. PATHOLOGY RESEARCH INTERNATIONAL 2010; 2011:903202. [PMID: 21188214 PMCID: PMC3005907 DOI: 10.4061/2011/903202] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 10/22/2010] [Indexed: 01/29/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is an important prognostic and predictive factor in breast cancer. HER2 is overexpressed in approximately 15%–20% of invasive breast carcinomas and is associated with earlier recurrence, shortened disease free survival, and poor prognosis. Trastuzumab (Herceptin) a “humanized” monoclonal antibody targets the extracellular domain of HER2 and is widely used in the management of HER2 positive breast cancers. Accurate assessment of HER2 is thus critical in the management of breast cancer. The aim of this paper is to present a comprehensive review of HER2 with reference to its discovery and biology, clinical significance, prognostic value, targeted therapy, current and new testing modalities, and the interpretation guidelines and pitfalls.
Collapse
Affiliation(s)
- Sejal Shah
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
796
|
Replication timing aberrations and aneuploidy in peripheral blood lymphocytes of breast cancer patients. Neoplasia 2010; 12:668-74. [PMID: 20689761 DOI: 10.1593/neo.10568] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Revised: 05/23/2010] [Accepted: 05/27/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Peripheral blood lymphocytes of patients with hematological malignancies or solid tumors, such as renal cell carcinoma or prostate cancer, display epigenetic aberrations (loss of synchronous replication of allelic counterparts) and genetic changes (aneuploidy) characteristic of the cancerous phenotype. This study sought to determine whether such alterations could differentiate breast cancer patients from cancer-free subjects. METHODS The HER2 locus-an oncogene assigned to chromosome 17 whose amplification is associated with breast cancer (BCA)-and the pericentromeric satellite sequence of chromosome 17 (CEN17) were used for replication timing assessments. Aneuploidy was monitored by enumerating the copy numbers of chromosome 17. Replication timing and aneuploidy were detected cytogenetically using fluorescence in situ hybridization technology applied to phytohemagglutinin-stimulated lymphocytes of 20 women with BCA and 10 control subjects. RESULTS We showed that both the HER2 and CEN17 loci in the stimulated BCA lymphocytes altered their characteristic pattern of synchronous replication and exhibited asynchronicity. In addition, there was an increase in chromosome 17 aneuploidy. The frequency of cells displaying asynchronous replication in the patients' samples was significantly higher (P < 10(-12) for HER2 and P < 10(-6) for CEN17) than the corresponding values in the control samples. Similarly, aneuploidy in patients' cells was significantly higher (P < 10(-9)) than that in the controls. CONCLUSIONS The HER2 and CEN17 aberrant replication differentiated clearly between BCA patients and control subjects. Thus, monitoring the replication of these genes offers potential blood markers for the detection and monitoring of breast cancer.
Collapse
|
797
|
The Role of Human Epidermal Growth Factor Receptor 2 as a Prognostic Factor in Lung Cancer: A Meta-Analysis of Published Data. J Thorac Oncol 2010; 5:1922-32. [PMID: 21155183 DOI: 10.1097/jto.0b013e3181f26266] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
798
|
Chen L, Placone J, Novicky L, Hristova K. The extracellular domain of fibroblast growth factor receptor 3 inhibits ligand-independent dimerization. Sci Signal 2010; 3:ra86. [PMID: 21119106 DOI: 10.1126/scisignal.2001195] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Dysregulation of the ligand-independent dimerization of receptor tyrosine kinases (RTKs), which is the first step in the activation of RTKs, leads to various pathologies. A mechanistic understanding of the dimerization process is lacking, and this lack of basic knowledge is one bottleneck in the development of effective RTK-targeted therapies. For example, the roles and relative contributions of the different domains of RTKs to receptor dimerization are unknown. Here, we used quantitative imaging Förster resonance energy transfer (QI-FRET) to determine the contribution of the extracellular domain of fibroblast growth factor receptor 3 (FGFR3) to the dimerization of the receptor. We provide evidence that the contribution of the extracellular domain of FGFR3 to dimerization is repulsive in the absence of ligand and is on the order of ~1 kcal/mol. The repulsive contribution of the extracellular domain is similar in magnitude, but opposite in sign, to the contribution of pathogenic single-amino acid mutations to RTK signaling, and is therefore likely to be important for biological function. Together, these results highlight the fine balance in the domain interactions that regulate RTK dimerization and signaling.
Collapse
Affiliation(s)
- Lirong Chen
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | | |
Collapse
|
799
|
Musolino A, Ciccolallo L, Panebianco M, Fontana E, Zanoni D, Bozzetti C, Michiara M, Silini EM, Ardizzoni A. Multifactorial central nervous system recurrence susceptibility in patients with HER2-positive breast cancer: epidemiological and clinical data from a population-based cancer registry study. Cancer 2010; 117:1837-46. [PMID: 21509760 DOI: 10.1002/cncr.25771] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 09/28/2010] [Accepted: 10/05/2010] [Indexed: 11/07/2022]
Abstract
BACKGROUND A series of retrospective studies have reported that patients with human epidermal growth factor receptor 2(HER2)-positive breast cancer are at a greater risk of central nervous system (CNS) metastases. Trastuzumab, which does not cross the blood-brain barrier, has been associated with this increased risk. METHODS The authors evaluated incidence, survival, and risk factors for CNS metastases in the incident breast cancer population systematically collected by the Parma Province Cancer Registry over the 4-year period between 2004 and 2007. RESULTS A total of 1458 patients with a diagnosis of stage I to III invasive breast cancer were analyzed for study purposes. At a median follow-up of 4.1 years, CNS events were observed in 1.3% and 5% of HER2-negative patients and HER2-positive patients, respectively (P < .0001). The administration of trastuzumab either as adjuvant therapy or for metastatic disease was associated with a significantly increased risk of CNS involvement at first disease recurrence and after first extracranial recurrence, respectively. According to multivariate analysis, HER2-positive status and trastuzumab treatment, high Ki-67 index, and hormone receptor negativity remained independent risk factors for the development of CNS metastasis. CONCLUSIONS To the authors' knowledge, this is the first population-based cancer registry study analyzing factors associated with CNS recurrence in a general population of newly diagnosed breast cancer patients with known HER2 status. The data from the current study provide evidence that patients with HER2-positive breast cancer have a significantly higher incidence of CNS metastasis after treatment with trastuzumab. Improvements in systemic control and overall survival associated with trastuzumab-based therapy may lead to an "unmasking" of CNS disease recurrence that would otherwise remain clinically silent before a patient's death.
Collapse
Affiliation(s)
- Antonino Musolino
- Medical Oncology Unit and Cancer Registry of Parma Province, University Hospital of Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
800
|
Baxevanis CN, Voutsas IF, Gritzapis AD, Perez SA, Papamichail M. HER-2/neu as a target for cancer vaccines. Immunotherapy 2010; 2:213-26. [PMID: 20635929 DOI: 10.2217/imt.09.89] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A novel modality toward the treatment of HER-2/neu-positive malignancies, mostly including breast and, more recently prostate carcinomas, has been the use of vaccines targeting HER-2/neu extracellular and intracellular domains. HER-2/neu-specific vaccines have been demonstrated to generate durable T-cell anti-HER-2/neu immunity when tested in Phase I and II clinical trials with no significant toxicity or autoimmunity directed against normal tissues. Targeting of HER-2/neu in active immunotherapy may involve peptide and DNA vaccines. Moreover, active anti-HER-2/neu immunization could facilitate the ex vivo expansion of HER-2/neu-specific T cells for use in adoptive immunotherapy for the treatment of established metastatic disease. In addition, early data from trials examining the potential use of HER-2/neu-based vaccines in the adjuvant setting to prevent the relapse of breast cancer in high-risk patients have shown promising results. Future approaches include multiepitope preventive vaccines and combinatorial treatments for generating the most efficient protective anti-tumor immunity.
Collapse
|