Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Front Oncol ; 11: 676939, 2021.
Article in English | MEDLINE | ID: mdl-34367962

ABSTRACT

The development of trastuzumab (Herceptin®) was one of the most significant cancer drug development projects of the 20th century. Not only was it a scientific and medical achievement but it also paved the way for the drug-diagnostic codevelopment model, where a predictive biomarker assay is developed in parallel to the drug. One of the challenges in the development of trastuzumab was to select the right patient population likely to respond and here, it was critical to have access to an accurate, robust and reliable assay for detection of HER2 overexpression in tumors. In the clinical development of trastuzumab, a clinical trial assay (CTA), developed by Genentech, was used for selection of HER2 positive patients. However, during the phase III trial with trastuzumab, a new optimized IHC assay, HercepTest™ was designed and developed by Dako. In the final stage of its development, a comparative study with the CTA was conducted in order to show concordance between the two assays. In September 1998, the Food and Drug Administration (FDA) simultaneously granted approval to trastuzumab and HercepTest™. The assay has been used for patient selection in a number of significant breast cancer clinical trials such as the HERA, CLEOPATRA, EMILIA and more. In these trials, HercepTest™ demonstrated its clinical utility in the neoadjuvant, adjuvant, and metastatic setting as well as in relation to different types of HER2 targeted therapies. Likewise, the assay was used for selection of HER2 positive gastric cancer patients in the important ToGA trail. HercepTest™ was the first companion diagnostic ever approved by the FDA, and more than 20 years of use has documented its clinical impact.

2.
PLoS One ; 13(3): e0194609, 2018.
Article in English | MEDLINE | ID: mdl-29566065

ABSTRACT

INTRODUCTION: Effective predictive biomarkers for selection of patients benefiting from adjuvant platinum-based chemotherapy in non-small cell lung cancer (NSCLC) are needed. Based on a previously validated methodology, molecular profiles of predicted sensitivity in two patient cohorts are presented. METHODS: The profiles are correlations between in vitro sensitivity to cisplatin and vinorelbine and baseline mRNA expression of the 60 cell lines in the National Cancer Institute panel. An applied clinical samples filter focused the profiles to clinically relevant genes. The profiles were tested on 1) snap-frozen tumors from 133 patients with completely resected stage 1B-2 NSCLC randomized to adjuvant cisplatin and vinorelbine (ACV, n = 71) or no adjuvant treatment (OBS, n = 62) and 2) formalin-fixed paraffin-embedded (FFPE) tumors from 95 patients with completely resected stage 1A-3B NSCLC receiving adjuvant cisplatin and vinorelbine. RESULTS: The combined cisplatin and vinorelbine profiles showed: 1) univariate Hazard Ratio (HR) for sensitive versus resistant of 0.265 (95% CI:0.079-0.889, p = 0.032) in the ACV cohort and a HR of 0.28 in a multivariate model (95% CI:0.08-1.04, p = 0.0573); 2) significant prediction at 3 year survival from surgery in univariate (HR = 0.138 (95% CI:0.035-0.537), p = 0.004) and multivariate analysis (HR = 0.14 (95% CI:0.030-0.6), p = 0.0081). No discrimination was found in the OBS cohort (HR = 1.328, p = 0.60). The cisplatin predictor alone had similar figures with 1) univariate HR of 0.37 (95% CI:0.12-1.15, p = 0.09) in the ACV cohort and 2) univariate HR of 0.14 (95% CI:0.03-0.59, p = 0.0076) to three years. Functional analysis on the cisplatin profile revealed a group of upregulated genes related to RNA splicing as a part of DNA damage repair and apoptosis. CONCLUSIONS: Profiles derived from snap-frozen and FFPE NSCLC tissue were prognostic and predictive in the patients that received cisplatin and vinorelbine but not in the cohort that did not receive adjuvant treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Molecular Diagnostic Techniques/methods , Transcriptome , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line, Tumor , Chemotherapy, Adjuvant , Cohort Studies , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/surgery , Male , Middle Aged , Prognosis , Randomized Controlled Trials as Topic , Treatment Outcome
3.
J Natl Cancer Inst ; 105(17): 1284-91, 2013 Sep 04.
Article in English | MEDLINE | ID: mdl-23964133

ABSTRACT

BACKGROUND: Methods using cell line microarray and drug sensitivity data to predict patients' chemotherapy response are appealing, but groups may be reluctant to release details to preserve intellectual property. Here we describe a case study to validate predictions while treating the methods as a "black box." METHODS: Medical Prognosis Institute (MPI) constructed cell-line-derived sensitivity scores (SSs) and combined scores (CSs) that incorporate clinical variables. MD Anderson researchers evaluated their predictions. We searched the Gene Expression Omnibus (GEO) to identify validation datasets, and we performed statistical evaluation of the agreement between prediction and clinical observation. RESULTS: We identified 3 suitable datasets: GSE16446 (n = 120; binary outcome), GSE17920 (n = 130; binary outcome), and GSE10255 (n = 161; continuous and time-to-event outcomes). The SS was statistically significantly associated with primary treatment responses for all studies (GSE16446: P = .02; GSE17920: P = .02; GSE10255: P = .02). Dichotomized SSs performed no better than chance for GSE16446 and GSE17920, and categorized SSs did not predict disease-free survival (GSE10255). SSs sometimes improved on predictions using clinical variables (GSE16446: P = .05; GSE17920: P = .31; GSE10255: P = .045), but gains were limited (95% confidence intervals for GSE16446 and GSE17920 include 0). The CS did not predict treatment response for GSE16446 (P = .55), but it did for GSE17920 (P < .001). Coefficients of clinical variables provided by MPI for CSs agree with estimates for GSE17920 better than estimates for GSE16446. CONCLUSIONS: Model predictions were better than chance in all three datasets. However, these scores added little to existing clinical predictors; statistically significant contributions were likely to be too small to change clinical practice. These findings suggest that discovering better predictors will require both cell line data and a clinical training dataset of patient samples.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor/drug effects , Hodgkin Disease/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Area Under Curve , Bleomycin/administration & dosage , Dacarbazine/administration & dosage , Doxorubicin/administration & dosage , Epirubicin/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Humans , Linear Models , Logistic Models , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , ROC Curve , Treatment Outcome , Vinblastine/administration & dosage
4.
Per Med ; 8(2): 137-148, 2011 Mar.
Article in English | MEDLINE | ID: mdl-29783414

ABSTRACT

The clinical utility and medico-economic value of several personalized diagnostic tests has been well described in the literature. Development of such tests, including generation of the necessary supportive clinical validation data, is a complex and expensive endeavor. In general, sponsors of such tests lack sufficient clarity on appropriate reimbursement and regulatory pathways to provide the clear development framework necessary to incentivize the required level of investment. In the USA, an imperfect reimbursement paradigm has evolved to accommodate a small number of 'value-priced' laboratory-developed tests, although major structural barriers remain to broader implementation. In Europe, by contrast, there is virtually no precedent for value-based public sector pricing, and even such procedurally based pricing as currently exists is administered by a complex network of largely decentralized bodies. As a consequence, patient access is limited and health-economic savings are not realized. This article explores some of the European market entry barriers, with a focus on reimbursement challenges, and highlights some collaborative proposals to address such.

5.
Appl Immunohistochem Mol Morphol ; 15(3): 325-31, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721279

ABSTRACT

Estrogen receptor (ER) and progesterone receptor (PR) status in breast carcinomas are considered validated predictive factors for selecting patients for antihormonal therapy. Published surveys have shown a significant rate of disagreement and lack of reproducibility of immunohistochemistry (IHC) results from laboratories around the world. To address these limitations IHC assays for ER and PR were developed using characterized reagents, after careful calibration of the sensitivity and specificity to match established assays previously validated in large clinical studies. The ER assay uses a cocktail of 2 mouse monoclonal antibodies (1D5 and ER-2-123) and the PR assay uses 1 mouse monoclonal antibody (PgR 1294); both are followed by a polymer-peroxidase-based detection system. All antibodies were tested for specificity by epitope mapping. The sensitivity of the new assays was calibrated to be equivalent to previously validated IHC assays followed by a comparison with the validated assays in a concordance study involving over 200 specimens. All slides were scored with the "Allred Score," also used for scoring of the original validated assays. The overall concordance between the new and the established IHC assays was nearly perfect (99%). The concordance study demonstrated greater than 98% positive agreement and 100% negative agreement of the new IHC assays with the previously validated IHC assays. This equivalence establishes the clinical validation of the assays and, as they are based on newer generation reagents and are produced and tested under stringent quality control conditions to ensure their consistency, they add additional advantages to the user and patients.


Subject(s)
Breast Neoplasms/diagnosis , Carcinoma/diagnosis , Immunohistochemistry/methods , Neoplasms, Hormone-Dependent/diagnosis , Patient Selection , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Animals , Antibodies, Monoclonal/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/pathology , Female , Humans , Mice , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology
6.
Prostate ; 60(3): 246-56, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15176054

ABSTRACT

BACKGROUND: Prostein is a recently described molecule expressed at the mRNA level in a prostate-specific manner. A murine monoclonal antibody was developed, characterized, and used to evaluate the expression of prostein protein in prostatic, other normal tissue and tumor samples. METHODS: The murine anti-prostein monoclonal antibody 10E3-G4-D3 was generated using recombinant prostein. ELISA, FACS, and Western analyzes were used to characterize 10E3-G4-D3. Immunohistochemistry was used to characterize the expression of prostein in tissues. RESULTS: 10E3-G4-D3 specifically recognizes a linear intracellular epitope of prostein. IHC analysis demonstrates that prostein is expressed in the vast majority of normal and malignant prostatic tissues, regardless of grade and metastatic status. No protein expression is detected in a panel of approximately 4,700 normal and malignant tissue samples representing the great majority of essential tissues and tumors. CONCLUSIONS: Prostein is exquisitely specific for prostate tissues, indicating a potential clinical utility of 10E3-G4-D3 as a diagnostic biomarker, and support the use of prostein as a novel target for development of prostein-specific antibody and T-cell based therapeutic strategies for prostate cancer.


Subject(s)
Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic , Membrane Proteins/biosynthesis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Antibodies, Monoclonal , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Mice , Neoplasms/genetics , Neoplasms/pathology , RNA, Messenger/analysis , Tissue Distribution , Tumor Cells, Cultured
7.
J Leukoc Biol ; 76(2): 406-15, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15136585

ABSTRACT

Serglycin is a major proteoglycan of hematopoietic cells. It is thought to play a role in the packaging of granule proteins in human neutrophil granulocytes. The presence of serglycin in myeloid cells has been demonstrated only at the transcriptional level. We generated a polyclonal antibody against recombinant human serglycin. Here, we show the localization of serglycin in humans during neutrophil differentiation. Immunocytochemistry revealed serglycin immunoreactivity in the Golgi area of promyelocytes (PM) and myelocytes (MC), as well as in a few band cells and mature neutrophil granulocytes. Granular staining was detected near the Golgi apparatus in some of the PM, and the major part of the cytoplasm was negative. Immunoelectron microscopy showed serglycin immunoreactivity located to the Golgi apparatus and a few immature granules of PM and MC. The decreasing level of serglycin protein during myeloid differentiation coincided with a decrease of mRNA expression, as evaluated by Northern blotting. Subcellular fractions of neutrophil granulocytes were obtained. Serglycin immunoreactivity was detected in the fraction containing Golgi apparatus, plasma membrane, and secretory vesicles by Western blotting and enzyme-linked immunosorbent assay. Serglycin was not detected in subcellular fractions containing primary, secondary, or tertiary granules. Together, these findings indicate that serglycin is located to the Golgi apparatus and a few immature granules during neutrophil differentiation. This is consistent with a function for serglycin in formation of granules in neutrophil granulocytes. Our findings contrast the view that native serglycin is present in mature granules and plays a role in packaging and regulating the activity of proteolytic enzymes there.


Subject(s)
Neutrophils/metabolism , Proteoglycans/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Microscopy, Immunoelectron , Neutrophils/immunology , Neutrophils/ultrastructure , Proteoglycans/genetics , Proteoglycans/immunology , RNA, Messenger/metabolism , Vesicular Transport Proteins
8.
Int J Radiat Oncol Biol Phys ; 58(2): 561-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14751528

ABSTRACT

Accelerated repopulation in head-and-neck carcinomas might be related to the expression of proliferative factors such as epidermal growth factor receptor (EGFr). The present study focuses on the prognostic value of EGFr for T-site control and the relation to tumor cell differentiation and overall treatment time. We studied 336 patients treated with primary radiotherapy using 66-68 Gy, 2 Gy per fraction and overall treatment times of 912, 612, or 512 weeks. Pretreatment biopsies were stained for EGFr.Thirty-five percent of the carcinomas had less than 50% of the area stained for EGFr. Small T-size and well-differentiated tumors was associated with a high degree of staining (p = 0.001 and p = 0.002, respectively). EGFr was of poor prognostic influence regarding local control in patients treated with 9 weeks split-course, whereas the opposite was found for patients given accelerated treatment in 5 weeks. A similar relationship between outcome, overall treatment time, and differentiation has previously been shown. The two parameters were analyzed together by separating the tumors with low EGFr and/or poor differentiation from tumors with well/moderate differentiation and high EGFr, resulting in odds ratios for T-site failure of 12 (1.43-104), 0.91 (0.51-1.65), and 0.43 (0.17-1.08), for treatment times of 912, 612, and 512 weeks, respectively. The tumor response to variations in fractionation is heterogeneous, and the prognostic impact of EGFr and differentiation might be relative and dependent on the overall treatment time of radiotherapy.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Neoplasm Proteins/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Head and Neck Neoplasms/pathology , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Laryngeal Neoplasms/radiotherapy , Pharyngeal Neoplasms/metabolism , Pharyngeal Neoplasms/pathology , Pharyngeal Neoplasms/radiotherapy , Prognosis , Radiotherapy Dosage , Randomized Controlled Trials as Topic , Statistics as Topic
9.
Biochim Biophys Acta ; 1591(1-3): 29-35, 2002 Aug 19.
Article in English | MEDLINE | ID: mdl-12183052

ABSTRACT

The various granule subtypes of the human neutrophil differ in propensity for exocytosis. As a rule, granules formed at late stages of myelopoiesis have a higher secretory potential than granules formed in more immature myeloid cells. Neutrophils contain four closely related alpha-defensins, which are stored in a subset of azurophil granules. These defensin-rich azurophil granules (DRG) are formed later than defensin-poor azurophil granules, near the promyelocyte/myelocyte transition. In order to characterize the secretory properties of DRG, we developed a sensitive and accurate ELISA for detection of the neutrophil alpha-defensins HNP 1-3. This allowed us to quantify the exocytosis of alpha-defensins and markers of azurophil (myeloperoxidase), specific (lactoferrin) and gelatinase (gelatinase) granules from neutrophils stimulated with different secretagogues. The release pattern of alpha-defensins correlated perfectly with the release of myeloperoxidase and showed no resemblance to the exocytosis of lactoferrin or gelatinase. This finding was substantiated through subcellular fractionation experiments. In conclusion, despite a distinct profile of biosynthesis, DRG are indistinguishable from defensin-poor azurophil granules with respect to exocytosis. Thus, in contrast to peroxidase-negative granules, azurophil granules display homogeneity in their availability for extracellular release.


Subject(s)
Defensins/metabolism , Neutrophils/metabolism , Antibodies , Cytochalasin B/pharmacology , Defensins/immunology , Enzyme-Linked Immunosorbent Assay/methods , Exocytosis , Humans , In Vitro Techniques , Ionomycin/pharmacology , Ionophores/pharmacology , Neutrophils/drug effects , Reproducibility of Results , Subcellular Fractions
SELECTION OF CITATIONS
SEARCH DETAIL