Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Lancet ; 389(10064): 67-76, 2017 01 07.
Article in English | MEDLINE | ID: mdl-27939400

ABSTRACT

BACKGROUND: First-line chemotherapy for patients with cisplatin-ineligible locally advanced or metastatic urothelial carcinoma is associated with short response duration, poor survival, and high toxicity. This study assessed atezolizumab (anti-programmed death-ligand 1 [PD-L1]) as treatment for metastatic urothelial cancer in cisplatin-ineligible patients. METHODS: For this single-arm, multicentre, phase 2 study, in 47 academic medical centres and community oncology practices in seven countries in North America and Europe, we recruited previously untreated patients with locally advanced or metastatic urothelial cancer who were cisplatin ineligible. Patients were given 1200 mg intravenous atezolizumab every 21 days until progression. The primary endpoint was independently confirmed objective response rate per Response Evaluation Criteria in Solid Tumors version 1.1 (central review), assessed in prespecified subgroups based on PD-L1 expression and in all patients. All participants who received one or more doses of atezolizumab were included in the primary and safety analyses. This study was registered with ClinicalTrials.gov, number NCT02108652. FINDINGS: Between June 9, 2014, and March 30, 2015, we enrolled 123 patients, of whom 119 received one or more doses of atezolizumab. At 17·2 months' median follow-up, the objective response rate was 23% (95% CI 16 to 31), the complete response rate was 9% (n=11), and 19 of 27 responses were ongoing. Median response duration was not reached. Responses occurred across all PD-L1 and poor prognostic factor subgroups. Median progression-free survival was 2·7 months (2·1 to 4·2). Median overall survival was 15·9 months (10·4 to not estimable). Tumour mutation load was associated with response. Treatment-related adverse events that occurred in 10% or more of patients were fatigue (36 [30%] patients), diarrhoea (14 [12%] patients), and pruritus (13 [11%] patients). One treatment-related death (sepsis) occurred. Nine (8%) patients had an adverse event leading to treatment discontinuation. Immune-mediated events occurred in 14 (12%) patients. INTERPRETATION: Atezolizumab showed encouraging durable response rates, survival, and tolerability, supporting its therapeutic use in untreated metastatic urothelial cancer. FUNDING: F Hoffmann-La Roche, Genentech.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/secondary , Urologic Neoplasms/drug therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , B7-H1 Antigen/blood , Biomarkers, Tumor/blood , Carcinoma, Transitional Cell/blood , Cisplatin , Contraindications , Female , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Response Evaluation Criteria in Solid Tumors , Urologic Neoplasms/blood
2.
Lancet ; 387(10031): 1909-20, 2016 May 07.
Article in English | MEDLINE | ID: mdl-26952546

ABSTRACT

BACKGROUND: Patients with metastatic urothelial carcinoma have few treatment options after failure of platinum-based chemotherapy. In this trial, we assessed treatment with atezolizumab, an engineered humanised immunoglobulin G1 monoclonal antibody that binds selectively to programmed death ligand 1 (PD-L1), in this patient population. METHODS: For this multicentre, single-arm, two-cohort, phase 2 trial, patients (aged ≥18 years) with inoperable locally advanced or metastatic urothelial carcinoma whose disease had progressed after previous platinum-based chemotherapy were enrolled from 70 major academic medical centres and community oncology practices in Europe and North America. Key inclusion criteria for enrolment were Eastern Cooperative Oncology Group performance status of 0 or 1, measurable disease defined by Response Evaluation Criteria In Solid Tumors version 1.1 (RECIST v1.1), adequate haematological and end-organ function, and no autoimmune disease or active infections. Formalin-fixed paraffin-embedded tumour specimens with sufficient viable tumour content were needed from all patients before enrolment. Patients received treatment with intravenous atezolizumab (1200 mg, given every 3 weeks). PD-L1 expression on tumour-infiltrating immune cells (ICs) was assessed prospectively by immunohistochemistry. The co-primary endpoints were the independent review facility-assessed objective response rate according to RECIST v1.1 and the investigator-assessed objective response rate according to immune-modified RECIST, analysed by intention to treat. A hierarchical testing procedure was used to assess whether the objective response rate was significantly higher than the historical control rate of 10% at an α level of 0·05. This study is registered with ClinicalTrials.gov, number NCT02108652. FINDINGS: Between May 13, 2014, and Nov 19, 2014, 486 patients were screened and 315 patients were enrolled into the study. Of these patients, 310 received atezolizumab treatment (five enrolled patients later did not meet eligibility criteria and were not dosed with study drug). The PD-L1 expression status on infiltrating immune cells (ICs) in the tumour microenvironment was defined by the percentage of PD-L1-positive immune cells: IC0 (<1%), IC1 (≥1% but <5%), and IC2/3 (≥5%). The primary analysis (data cutoff May 5, 2015) showed that compared with a historical control overall response rate of 10%, treatment with atezolizumab resulted in a significantly improved RECIST v1.1 objective response rate for each prespecified immune cell group (IC2/3: 27% [95% CI 19-37], p<0·0001; IC1/2/3: 18% [13-24], p=0·0004) and in all patients (15% [11-20], p=0·0058). With longer follow-up (data cutoff Sept 14, 2015), by independent review, objective response rates were 26% (95% CI 18-36) in the IC2/3 group, 18% (13-24) in the IC1/2/3 group, and 15% (11-19) overall in all 310 patients. With a median follow-up of 11·7 months (95% CI 11·4-12·2), ongoing responses were recorded in 38 (84%) of 45 responders. Exploratory analyses showed The Cancer Genome Atlas (TCGA) subtypes and mutation load to be independently predictive for response to atezolizumab. Grade 3-4 treatment-related adverse events, of which fatigue was the most common (five patients [2%]), occurred in 50 (16%) of 310 treated patients. Grade 3-4 immune-mediated adverse events occurred in 15 (5%) of 310 treated patients, with pneumonitis, increased aspartate aminotransferase, increased alanine aminotransferase, rash, and dyspnoea being the most common. No treatment-related deaths occurred during the study. INTERPRETATION: Atezolizumab showed durable activity and good tolerability in this patient population. Increased levels of PD-L1 expression on immune cells were associated with increased response. This report is the first to show the association of TCGA subtypes with response to immune checkpoint inhibition and to show the importance of mutation load as a biomarker of response to this class of agents in advanced urothelial carcinoma. FUNDING: F Hoffmann-La Roche Ltd.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , Urologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/immunology , Carboplatin/administration & dosage , Cisplatin/administration & dosage , Disease Progression , Drug Administration Schedule , Female , Humans , Immunoglobulins, Intravenous/administration & dosage , Infusions, Intravenous , Male , Middle Aged , Mutation/genetics , Neoplasm Metastasis , Response Evaluation Criteria in Solid Tumors , Treatment Outcome , Urologic Neoplasms/genetics
3.
J Manag Care Spec Pharm ; 21(10): 863-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26402387

ABSTRACT

BACKGROUND: Stage IV breast cancer, also known as metastatic breast cancer (mBC), is not a curable condition. However, treatment can prolong life, delay the progression of the cancer, or improve quality of life. Currently, patients with mBC are often treated with chemotherapy. Patients often experience adverse events from chemotherapy during the treatment cycle, which leads to chemotherapy modifications such as dose delay, dose reduction, or discontinuation of chemotherapy. Previous studies have evaluated the rates of adverse events that occur from the use of chemotherapy; however, few studies have evaluated the clinical impact on the chemotherapy regimen once the adverse event occurs. This study evaluates the clinical impact on the chemotherapy regimen from chemotherapy-related adverse events in patients with mBC in an integrated health care delivery system.   OBJECTIVES: To assess the adverse events in patients with mBC and evaluate the clinical impact on the chemotherapy regimen from these adverse events in an integrated health care delivery system.  METHODS: This study is a retrospective cohort of patients with mBC newly initiated on chemotherapy. The first infusion was defined as the index date. Patients were aged greater than 18 years at time of index date and had 6 months or more of Kaiser membership and drug eligibility prior to the index date and continuous membership and drug eligibility throughout follow-up. Adverse events were identified after the index date and during the follow-up using ICD-9-CM diagnosis and procedure codes. Single or multiple episodes of care were created from the adverse events. Chart review was conducted to establish whether the adverse event was related to chemotherapy and if any modification to the chemotherapy regimen occurred-a dose delay, dose reduction, or discontinuation was considered a clinical impact on therapy. Multivariate logistic regression was used to examine factors associated with clinical impact versus no clinical impact from the delivery of chemotherapy treatment.  RESULTS: A total of 1,682 patients with mBC were identified during our time period with an average follow-up of 2.21 years on first-line chemotherapy (SD = 1.83). 909 patients (54%) had at least 1 adverse event, and 773 patients (46%) did not have any adverse events during follow-up. Significant differences at baseline between these 2 groups included race, peripheral vascular disease, and length of stay (P less than 0.05). From the 909 patients who had at least 1 adverse event, 185 patients (20%) experienced an impact on their chemotherapy regimens. Patients with single episodes of care with any chemotherapy regimen impact experienced mostly hematological, infection/pyrexia, and gastrointestinal-related adverse events. In multiple episodes of care, neurological impact was more frequent than gastrointestinal-related effects. Patients with hospitalizations of greater than 3 days experienced the most impact, demonstrating that severe adverse events have more impact on chemotherapy regimens. In our multivariate analysis, patients aged greater than 65 years, having more than 1 comorbidity and having longer duration in days for each episode of care were all associated with clinical impact. Black and Hispanic patients were more likely to have a modification in their chemotherapy compared with white patients. CONCLUSIONS: This retrospective analysis demonstrates that chemotherapy-related adverse events in patients with mBC have an impact on the delivery of chemotherapy regimens. Having multiple comorbidities, increased age, and prolonged hospitalizations because of adverse events appear to be some of the primary factors related to chemotherapy modification.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Delivery of Health Care, Integrated , Hospitalization/statistics & numerical data , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cohort Studies , Disease Progression , Female , Follow-Up Studies , Humans , Length of Stay , Logistic Models , Middle Aged , Multivariate Analysis , Neoplasm Staging , Quality of Life , Retrospective Studies
4.
Breast Cancer Res Treat ; 144(3): 673-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24596047

ABSTRACT

Treatment options for metastatic breast cancer (MBC) are complex, and some patients experience early discontinuation or switching of treatment (ETDS). We examined the relationship between ETDS and patient-reported symptom burden among patients receiving first-line treatment of MBC in community oncology settings. This retrospective observational study used the ACORN Data Warehouse, a comprehensive community oncology repository of medical records and patient-reported outcomes. Patients with first-line treatment for MBC who had Patient Care Monitor (PCM) surveys were eligible. ETDS was defined as: record stating ETDS, treatment duration < planned, and planned therapy <6 weeks. Symptom burden was measured by two PCM composite scores [continuous (0-22) and categorical (absent, mild, moderate, and severe)] computed from 22 PCM items with varying cut points to assess symptom burden over time. Cox regression with time-varying covariates was used to assess risk for ETDS controlling for patient characteristics and treatment type: chemo (chemotherapy without targeted therapy (±hormone therapy); targeted (chemotherapy plus targeted therapy (±hormone therapy); and hormone (hormone therapy only). Overall, 197 (24.7 %) of a total sample of 797 patients had an ETDS event, of which 109 (55.3 %) were switches rather than early discontinuation. ETDS rate was nominally lower in the hormone group (11.1 %) versus chemo (27.6 %) or targeted (26.1 %). PCM continuous composite score predicted ETDS, controlling for other variables (HR = 1.132, p < 0.0001). ETDS was predicted by moderate and severe levels of PCM categorical composite score (HR = 4.135, and HR = 5.287 vs. absent, respectively, both p < 0.0001), with the pattern suggesting a threshold effect. Moderate or severe levels of a wide range of patient-reported symptoms and the accumulation of symptoms over time significantly predicted ETDS. Providers may better maintain patients on planned therapy if they attend to overall symptom burden patients experience over time.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Substitution , Withholding Treatment , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/epidemiology , Female , Humans , Middle Aged , Neoplasm Metastasis , Retrospective Studies , Risk Factors , Survival Analysis , Treatment Outcome
5.
Breast Cancer Res Treat ; 141(3): 461-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24062208

ABSTRACT

Data characterizing demographics, treatment patterns, and clinical outcomes in black patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC) are limited. registHER is a large, observational cohort study of patients (n = 1,001) with HER2-positive MBC diagnosed ≤6 months of enrollment and followed until death, disenrollment, or June 2009 (median follow-up of 27 months). Demographics, treatment patterns, and clinical outcomes were described for black (n = 126) and white patients (n = 793). Progression-free survival (PFS) following first-line therapy and overall survival (OS) were examined. Multivariate analyses adjusted for baseline and treatment factors. Black patients were more likely than white patients to be obese (body mass index ≥30), to have diabetes, and to have a history of cardiovascular disease; they were also less likely to have estrogen receptor or progesterone receptor positive disease. In patients treated with trastuzumab, the incidence of cardiac safety events (grade ≥3) was higher in black patients (10.9 %) than in white patients (7.9 %). Unadjusted median OS and PFS (months) were significantly lower in black patients than in white patients (OS: black: 27.1, 95 % confidence interval [CI] 21.3-32.1; white: 37.3, 95 % CI 34.6-41.1; PFS: black: 7.0, 95 % CI 5.7-8.2; white: 10.2, 95 % CI 9.3-11.2). The adjusted OS hazard ratio (HR) for black patients compared with white patients was 1.29 (95 % CI 1.00-1.65); adjusted PFS HR was 1.29 (95 % CI 1.05-1.59). This real-world evaluation of a large cohort of patients with HER2-positive MBC shows poorer prognostic factors and independently worse clinical outcomes in black versus white patients. Further research is needed to identify potential biologic differences that could have predictive impact for black patients or that could explain these differences.


Subject(s)
Black or African American , Breast Neoplasms, Male/drug therapy , Breast Neoplasms/drug therapy , Healthcare Disparities , Receptor, ErbB-2/metabolism , White People , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms, Male/mortality , Breast Neoplasms, Male/pathology , Disease-Free Survival , Female , Heart Failure/chemically induced , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Proportional Hazards Models , Racism , Trastuzumab , Treatment Outcome , Young Adult
6.
Breast J ; 19(5): 470-7, 2013.
Article in English | MEDLINE | ID: mdl-23865786

ABSTRACT

The majority of clinical trials of neo-adjuvant therapy for breast cancer have been conducted in resource-rich countries. We chose Nigeria, a resource-poor country, as the major site for a phase II feasibility open-label multicenter clinical trial designed to evaluate the efficacy, safety, and tolerability of neo-adjuvant capecitabine in locally advanced breast cancer (LABC). Planned treatment consisted of 24 weeks of capecitabine at a dose of 1,000 mg/m(2) twice daily (2,000 mg/m(2) total per day). The primary endpoints were overall, partial, complete clinical response rate (OCR, PCR, CCR) and complete pathologic response (cPR). A total of 16 patients were recruited from August 2007 to April 2010. The study was terminated early as a result of slow accrual. After the first three cycles of therapy, PCR were seen in five of 16 patients (31%; 95% CI 11-59%). Of the remaining 11 patients, eight had no response (NR) or stable disease (SD), and three had progressive disease (PD). Seven patients proceeded with further therapy of which had SD. OCR at the end of eight cycles was 44% (95% CI 20-70%). Clinical response and radiologic response by ultrasonomammography were highly concordant (spearman correlation 0.70). The most common adverse effect was Grade 1 hand-foot syndrome, which was seen in 75% of patients. Despite several limitations, we successfully carried out this phase II feasibility study of neo-adjuvant capecitabine for LABC in Nigeria. Capecitabine monotherapy showed good overall response rates with minimal toxicity and further studies are warranted.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Neoadjuvant Therapy , Adult , Aged , Capecitabine , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Feasibility Studies , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Humans , Middle Aged , Nigeria
7.
Lung Cancer ; 63(2): 169-79, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18672314

ABSTRACT

The receptor tyrosine kinase MET has been studied of a large variety of human cancers, including lung and mesothelioma. The MET receptor and its ligand HGF (hepatocyte growth factor) play important roles in cell growth, survival and migration, and dysregulation of the HGF-MET pathway leads to oncogenic changes including tumor proliferation, angiogenesis and metastasis. In small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), and malignant pleural mesothelioma (MPM), MET is dysregulated via overexpression, constitutive activation, gene amplification, ligand-dependent activation, mutation or epigenetic mechanisms. New drugs targeted against MET and HGF are currently being investigated in vitro and in vivo, with promising results. These drugs function at a variety of steps within the HGF-MET pathway, including MET expression at the RNA or protein level, the ligand-receptor interaction, and tyrosine kinase function. This paper will review the structure, function, mechanisms of tumorigenesis, and potential for therapeutic inhibition of the MET receptor in lung cancer and mesothelioma.


Subject(s)
Proto-Oncogene Proteins c-met/antagonists & inhibitors , Thoracic Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/etiology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/etiology , Mesothelioma/drug therapy , Mesothelioma/etiology , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/physiology
8.
ScientificWorldJournal ; 8: 909-19, 2008 Sep 21.
Article in English | MEDLINE | ID: mdl-18836658

ABSTRACT

Cancers of the head and neck and of the lung are associated with high morbidity and mortality rates that have remained relatively unchanged for more than 3 decades, despite advances in radiation therapies and chemotherapies over the same time. It is generally believed that the efficacy of standard therapy regimens has reached a plateau for these cancers. The discovery of specific aberrant molecular signaling pathways in solid tumors has afforded promising new directions for newer "targeted" cancer therapeutics. Among these, the epidermal growth factor receptor (EGFR) shows promise as a therapeutic target. Clinical studies have demonstrated that this targeted approach provides clinically meaningful benefit. This article reviews EGFR-targeted therapies in use and in development, with a focus on the role of EGFR in the pathophysiology of head and neck and lung cancer, and new concepts being investigated to improve outcomes with these agents.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/drug effects , Head and Neck Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized , Bevacizumab , Cetuximab , Clinical Trials as Topic , Combined Modality Therapy , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/immunology , Erlotinib Hydrochloride , Gefitinib , Humans , Lapatinib , Panitumumab , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use
9.
Nat Clin Pract Oncol ; 4(2): 118-29, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259932

ABSTRACT

Lung carcinoma is the most commonly diagnosed cancer and the leading cause of cancer deaths in the US. It accounts for 12% of all cancers diagnosed worldwide, making it the most common malignancy, other than nonmelanoma skin cancer. A new focus has emerged involving the role of race and ethnicity in lung carcinoma. Current health statistics data demonstrate striking disparities, which are most evident between African American patients and their white counterparts. This disparity is greatest among male patients, where statistically significant differences are seen not only in lung cancer incidence and risk, but also in survival and treatment outcomes. Several hypotheses that attempt to explain this disparity include genetic, cultural and socioeconomic differences, in addition to differences in tobacco use and exposure. Current evidence does not clearly identify the reasons for this observed disparity, or the role the aforementioned factors play in the development and overall outcomes of people with lung cancer in these populations. This disease continues to pose a considerable public health burden and more research is needed to improve understanding of the disparity of lung carcinoma statistics among African Americans. This review summarizes the existing body of knowledge regarding lung carcinoma in African Americans and attempts to identify promising areas for future investigation and intervention.


Subject(s)
Black or African American , Genetic Predisposition to Disease , Lung Neoplasms/ethnology , Lung Neoplasms/epidemiology , Black or African American/ethnology , Black or African American/genetics , Health Services Accessibility , Humans , Incidence , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mass Screening/statistics & numerical data , Prognosis , Risk Factors , Smoking/adverse effects , Social Class , Survival Analysis
10.
Rev Recent Clin Trials ; 2(2): 143-7, 2007 May.
Article in English | MEDLINE | ID: mdl-18473999

ABSTRACT

Receptor tyrosine kinases are a group of molecules that can enhance cellular proliferation, cell motility and migration, and eventual metastasis. c-Met receptor tyrosine kinase has a significant biological and biochemical effect on cancer cells, and appears to be an important therapeutic target. In many cancers, c-Met (which can be activated by its ligand hepatocyte growth factor, HGF) can be overexpressed, activated, amplified, and/or mutated. The mutations of c-Met had initially been described in the tyrosine kinase domain, and we have described them in other "hot-spots" such as the juxtamembrane and semaphorin domains. Targeting c-Met has been very fruitful pre-clinically, and currently, there are several clinical trials for advanced cancers. Described in this review are some of the biological and biochemical aspects of c-Met, and detailed are a number of therapeutic strategies. With our understanding of c-Met biology and role in cancer, we should be able to arrive at a unique strategy to eradicate cancers in which c-Met plays a significant role.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/metabolism , Clinical Trials as Topic , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...