Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Trials ; 19(5): 561-572, 2022 10.
Article in English | MEDLINE | ID: mdl-35786000

ABSTRACT

BACKGROUND/AIM: The number of coronavirus disease 2019 deaths and cases continues to increase globally. Novel therapies are urgently needed to treat patients with coronavirus disease 2019. We sought to provide a critical review of trials designed during the coronavirus disease 2019 pandemic. Our primary goal was to provide a critical review of the landscape of clinical trials designed to address the coronavirus disease 2019 pandemic. Specifically, we were interested in assessing the design of phase II/III and phase III interventional trials. METHODS: We utilized the ClinicalTrials.gov database to include trials registered between 1 December 2019 and 11 April 2021 to survey the current landscape of clinical trials for coronavirus disease 2019. Variables extracted included: National Clinical Trial number, title, location, sponsor, study type, start date, completion date, gender group, age group, primary outcome, secondary outcome, overall status, and associated references. RESULTS: About 57% of studies were interventional, 14.5% were phase III trials, and the majority of the therapeutic trials included hospitalized patients. There were 52 primary composite outcomes and 285 unique interventions spanning 10 drug classes. The outcomes, disease severity, and comparators varied substantially across trials, and the trials were often too small to be definitive. CONCLUSION: These findings are relevant as we strongly advocate for global coordination of efforts through the use of common platforms that enable harmonizing of endpoints, collection of common key variables and clear definition of disease severity to have clinically meaningful results from clinical trials.


Subject(s)
COVID-19 , Humans , Pandemics , Research Design , SARS-CoV-2 , Severity of Illness Index
2.
N Engl J Med ; 366(19): 1770-81, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22571201

ABSTRACT

BACKGROUND: Data are lacking on whether lenalidomide maintenance therapy prolongs the time to disease progression after autologous hematopoietic stem-cell transplantation in patients with multiple myeloma. METHODS: Between April 2005 and July 2009, we randomly assigned 460 patients who were younger than 71 years of age and had stable disease or a marginal, partial, or complete response 100 days after undergoing stem-cell transplantation to lenalidomide or placebo, which was administered until disease progression. The starting dose of lenalidomide was 10 mg per day (range, 5 to 15). RESULTS: The study-drug assignments were unblinded in 2009, when a planned interim analysis showed a significantly longer time to disease progression in the lenalidomide group. At unblinding, 20% of patients who received lenalidomide and 44% of patients who received placebo had progressive disease or had died (P<0.001); of the remaining 128 patients who received placebo and who did not have progressive disease, 86 crossed over to lenalidomide. At a median follow-up of 34 months, 86 of 231 patients who received lenalidomide (37%) and 132 of 229 patients who received placebo (58%) had disease progression or had died. The median time to progression was 46 months in the lenalidomide group and 27 months in the placebo group (P<0.001). A total of 35 patients who received lenalidomide (15%) and 53 patients who received placebo (23%) died (P=0.03). More grade 3 or 4 hematologic adverse events and grade 3 nonhematologic adverse events occurred in patients who received lenalidomide (P<0.001 for both comparisons). Second primary cancers occurred in 18 patients who received lenalidomide (8%) and 6 patients who received placebo (3%). CONCLUSIONS: Lenalidomide maintenance therapy, initiated at day 100 after hematopoietic stem-cell transplantation, was associated with more toxicity and second cancers but a significantly longer time to disease progression and significantly improved overall survival among patients with myeloma. (Funded by the National Cancer Institute; ClinicalTrials.gov number, NCT00114101.).


Subject(s)
Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Stem Cell Transplantation , Thalidomide/analogs & derivatives , Adult , Aged , Antineoplastic Agents/adverse effects , Disease-Free Survival , Double-Blind Method , Female , Follow-Up Studies , Humans , Lenalidomide , Maintenance Chemotherapy , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Neoplasms, Second Primary/epidemiology , Thalidomide/adverse effects , Thalidomide/therapeutic use
3.
J Neurooncol ; 120(1): 111-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25026995

ABSTRACT

The EORTC-NCIC regimen for glioblastoma requires different dosing of temozolomide (TMZ) during radiation and maintenance therapy. This complexity is exacerbated by the availability of multiple TMZ capsule strengths. TMZ is an alkylating agent and the major toxicity of this class is dose-related myelosuppression. Inadvertent overdose can be fatal. The websites of the Institute for Safe Medication Practices (ISMP), and the Food and Drug Administration (FDA) MedWatch database were reviewed. We searched the MedWatch database for adverse events associated with TMZ and obtained all reports including hematologic toxicity submitted from 1st November 1997 to 30th May 2012. The ISMP describes errors with TMZ resulting from the positioning of information on the label of the commercial product. The strength and quantity of capsules on the label were in close proximity to each other, and this has been changed by the manufacturer. MedWatch identified 45 medication errors. Patient errors were the most common, accounting for 21 or 47% of errors, followed by dispensing errors, which accounted for 13 or 29%. Seven reports or 16% were errors in the prescribing of TMZ. Reported outcomes ranged from reversible hematological adverse events (13%), to hospitalization for other adverse events (13%) or death (18%). Four error reports lacked detail and could not be categorized. Although the FDA issued a warning in 2003 regarding fatal medication errors and the product label warns of overdosing, errors in TMZ dosing occur for various reasons and involve both healthcare professionals and patients. Overdosing errors can be fatal.


Subject(s)
Adverse Drug Reaction Reporting Systems/statistics & numerical data , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Medication Errors/mortality , Brain Neoplasms/mortality , Dacarbazine/therapeutic use , Databases, Factual , Female , Follow-Up Studies , Glioblastoma/mortality , Humans , Male , Medication Errors/statistics & numerical data , Middle Aged , Pharmaceutical Preparations/administration & dosage , Prognosis , Retrospective Studies , Survival Rate , Temozolomide , United States
4.
JAMA ; 309(13): 1359-67, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23549581

ABSTRACT

IMPORTANCE: There are no known effective treatments for painful chemotherapy-induced peripheral neuropathy. OBJECTIVE: To determine the effect of duloxetine, 60 mg daily, on average pain severity. DESIGN, SETTING, AND PATIENTS: Randomized, double-blind, placebo-controlled crossover trial at 8 National Cancer Institute (NCI)-funded cooperative research networks that enrolled 231 patients who were 25 years or older being treated at community and academic settings between April 2008 and March 2011. Study follow-up was completed July 2012. Stratified by chemotherapeutic drug and comorbid pain risk, patients were randomized to receive either duloxetine followed by placebo or placebo followed by duloxetine. Eligibility required that patients have grade 1 or higher sensory neuropathy according to the NCI Common Terminology Criteria for Adverse Events and at least 4 on a scale of 0 to 10, representing average chemotherapy-induced pain, after paclitaxel, other taxane, or oxaliplatin treatment. INTERVENTIONS: The initial treatment consisted of taking 1 capsule daily of either 30 mg of duloxetine or placebo for the first week and 2 capsules of either 30 mg of duloxetine or placebo daily for 4 additional weeks. MAIN OUTCOME MEASURES: The primary hypothesis was that duloxetine would be more effective than placebo in decreasing chemotherapy-induced peripheral neuropathic pain. Pain severity was assessed using the Brief Pain Inventory-Short Form "average pain" item with 0 representing no pain and 10 representing as bad as can be imagined. RESULTS: Individuals receiving duloxetine as their initial 5-week treatment reported a mean decrease in average pain of 1.06 (95% CI, 0.72-1.40) vs 0.34 (95% CI, 0.01-0.66) among those who received placebo (P = .003; effect size, 0.513). The observed mean difference in the average pain score between duloxetine and placebo was 0.73 (95% CI, 0.26-1.20). Fifty-nine percent of those initially receiving duloxetine vs 38% of those initially receiving placebo reported decreased pain of any amount. CONCLUSION AND RELEVANCE: Among patients with painful chemotherapy-induced peripheral neuropathy, the use of duloxetine compared with placebo for 5 weeks resulted in a greater reduction in pain. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00489411.


Subject(s)
Antineoplastic Agents/adverse effects , Neuralgia/drug therapy , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Quality of Life , Selective Serotonin Reuptake Inhibitors/therapeutic use , Thiophenes/therapeutic use , Administration, Oral , Adult , Aged , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Duloxetine Hydrochloride , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Peripheral Nervous System Diseases/complications , Treatment Outcome
5.
J Neurooncol ; 100(1): 73-80, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20151173

ABSTRACT

Olfactory tumors, especially olfactory neuroblastomas (ON) and carcinomas with neuroendocrine differentiation (CND), are extremely rare, and little descriptive epidemiologic information is available. The objective of this study was to more fully describe selected olfactory tumors using a large population-based cancer incidence database. The Surveillance, Epidemiology and End Results (SEER) 9 registries limited-use data were reviewed from 1973 to 2006 for selected nasal cavity (C30.0) and accessory sinus (C31.0-31.9) tumors. Frequencies, incidence rates, and relative survival rates were estimated using SEER*Stat, v6.5.2. The majority of cases were squamous cell carcinoma (SCC), while the incidence of ON was greater than CND. For ON, the incidence was highest in the 60-79 year age group, while for SCC, the incidence was highest in the 80+ year age group. For CND, the incidence leveled off in the oldest age groups. Survival rates were highest for ON (>70% alive at 5 years after diagnosis) and poorest for CND (44% alive at 5 years). Adjuvant radiation therapy did not improve survival over surgery alone in ON. In SCC, survival was worse in patients who received adjuvant radiation compared to patients who had surgery alone. Our analysis confirms some previously published information, and adds new information about the incidence and demographics of ON and CND. In addition, our analysis documents the lack of benefit of adjuvant radiation in ON. It is not feasible to conduct prospective trials in patients with these rare diseases, and the importance of registry data in learning about olfactory tumors is emphasized.


Subject(s)
Brain Neoplasms/epidemiology , Carcinoma, Neuroendocrine/epidemiology , Neuroblastoma/epidemiology , Olfactory Pathways/pathology , Adolescent , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Carcinoma, Neuroendocrine/mortality , Child , Child, Preschool , Community Health Planning , Female , Humans , Incidence , Infant , Infant, Newborn , International Classification of Diseases , Male , Middle Aged , Neuroblastoma/mortality , Registries , SEER Program , Young Adult
6.
Ann Pharmacother ; 43(2): 235-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19193580

ABSTRACT

BACKGROUND: Serum creatinine (SCr)-based formulas are used to estimate glomerular filtration rate (GFR) when calculating a dosage for carboplatin using the Calvert equation, but these formulas often underestimate measured GFR. The Modified Diet in Renal Disease (MDRD) equation appears to be a more accurate estimate of GFR in patients with chronic kidney disease, but this equation has not been studied extensively in patients with cancer. OBJECTIVE: To determine the absolute difference between the dose of carboplatin administered (traditional SCr-based formulas used to estimate GFR) and the dose calculated using the MDRD equation to estimate GFR and compare the frequencies of thrombocytopenia, neutropenia, and dosage modifications between subjects in whom the difference in dose was 20% or more (divergent) or less than 20% (nondivergent). METHODS: A retrospective analysis was conducted using data from patients who received carboplatin. Inclusion criteria were receipt of at least 2 doses of carboplatin, either as monotherapy or combination therapy, and documentation of desired area under the concentration-time curve (AUC). Patients were excluded if the baseline values needed to estimate GFR using the MDRD equation were not available. The absolute difference between the dose of carboplatin administered and that calculated using the MDRD equation was determined and, from this comparison, the subjects were divided into 2 groups (divergent vs nondivergent). RESULTS: The medical records of 186 adults who received more than 2 doses of carboplatin were included in the analysis. The doses were divergent in 89 (48%) patients. The mean target AUC values were 5.3 mg/mL/min and 5.1 mg/mL/min in the divergent and nondivergent groups, respectively, and most patients received cytotoxic regimens with a relatively low risk of febrile neutropenia. The frequencies of neutropenia, thrombocytopenia, and dosage modifications were similar between the 2 groups. Use of the MDRD equation to calculate the carboplatin dosage did not appear to result in a change in the frequency of myelosuppression or the need for dose modifications compared with traditional SCr-based formulas. CONCLUSIONS: The traditional SCr-based formulas for the calculation of carboplatin dosage should be used to estimate carboplatin dose until more data become available regarding the use of the MDRD equation in this population.


Subject(s)
Carboplatin/administration & dosage , Drug Dosage Calculations , Carboplatin/adverse effects , Carboplatin/pharmacokinetics , Drug Therapy, Combination , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Neutrophils/drug effects , Platelet Count , Retrospective Studies
7.
Neuro Oncol ; 18(9): 1319-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27022131

ABSTRACT

BACKGROUND: p53 is a promising target in human cancer. p28 is a cell-penetrating peptide that preferentially enters cancer cells and binds to both wild-type and mutant p53 protein, inhibiting COP1-mediated ubiquitination and proteasomal degradation. This results in increased levels of p53, which induces cell cycle arrest at G2/M. We conducted a phase I study to determine the maximum-tolerated dose (MTD) and describe the dose-limiting toxicities (DLTs) and pharmacokinetics (PKs) of p28 in children. METHODS: Children aged 3-21 years with recurrent or progressive central nervous system tumors were eligible. Intravenous p28 was administered 3 times weekly for 4 consecutive weeks of a 6-week cycle at 4.16 mg/kg/dose (the adult recommended phase II dose) using a rolling-6 study design. Expression status of p53 was characterized by immunohistochemistry, and serum PK parameters were established on the second dose. RESULTS: Of the 18 eligible patients enrolled in the study, 12 completed the DLT monitoring period and were evaluable for toxicity. p28 was well-tolerated; 7 participants received ≥2 courses, and the most common adverse event attributed to the drug was transient grade 1 infusion-related reaction. PK analysis revealed a profile similar to adults; however, an increased area under the curve was observed in pediatric patients. High p53 expression in tumor cell nuclei was observed in 6 of 12 available tissue samples. There were no objective responses; 2 participants remained stable on the study for >4 cycles. CONCLUSIONS: This phase I study demonstrated that p28 is well-tolerated in children with recurrent CNS malignancies at the adult recommended phase II dose.


Subject(s)
Antineoplastic Agents/therapeutic use , Azurin/therapeutic use , Central Nervous System Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Peptide Fragments/therapeutic use , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects , Adolescent , Adult , Antineoplastic Agents/pharmacokinetics , Azurin/pharmacokinetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Peptide Fragments/pharmacokinetics , Prognosis , Young Adult
8.
CNS Drugs ; 29(11): 935-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26563196

ABSTRACT

The purpose of our review is to summarize the clinical activity of oral targeted agents against brain metastases. This includes BRAF inhibitors (dabrafenib and vemurafenib), human epidermal growth factor receptor inhibitors (lapatinib, gefitinib, erlotinib, and afatinib), multi-kinase angiogenesis inhibitors (sorafenib, sunitinib, pazopanib, and vandetanib), and ALK/c-MET (crizotinib) and ALK/IGF-1 (ceritinib) inhibitors. Effective systemic therapies are needed for long-term benefit in brain metastases and documentation of intracranial activity for many therapies is poor. Our review provides a summary of the literature with pertinent data for clinicians. This is needed as subjects with brain metastases are often prevented from enrolling in clinical trials and investigations focused on systemic therapies for brain metastases are rare.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Administration, Oral , Humans
9.
Cancer Chemother Pharmacol ; 71(6): 1561-5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23564377

ABSTRACT

BACKGROUND: Bevacizumab is widely used and may cause life-threatening bleeding. We attempted to identify clinical characteristics associated with central nervous system (CNS) hemorrhage in a broad population. METHODS: We performed a retrospective review of the FDA MedWatch database of adverse events reported with bevacizumab from 11/1997 to 5/2009. Our search used keywords: bleeding, hemorrhage, cerebral, intracranial, subarachnoid, cerebellar, hemorrhagic stroke and brain. RESULTS: A total of 17,466 reports were included in the database: 154 described CNS hemorrhage in 99 patients, and 1,041 reports described non-CNS bleeds. Median age was 62 years, and the primary cancers were consistent with indications for bevacizumab. Patients received a median of three (1-36) doses of bevacizumab prior to the bleed. Thirty percent had documented history of hypertension. Sixteen patients with CNS hemorrhage were reported to have CNS metastases. Death was reported as a complication of hemorrhage in 48 %. The most common predisposing factor for CNS bleeds was use of medications associated with bleeding, followed by thrombocytopenia. CONCLUSION: In this database, 154 of 1,195 reports of bleeding associated with bevacizumab described a CNS bleed. Although CNS bleeds were not common, they were the reported cause of death in two-thirds of cases.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Intracranial Hemorrhages/chemically induced , Neoplasms/drug therapy , Adverse Drug Reaction Reporting Systems , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Databases, Factual , Humans , Intracranial Hemorrhages/mortality , Risk Factors
10.
Cancer Chemother Pharmacol ; 69(4): 1113-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22147076

ABSTRACT

The benefit of six cycles of adjuvant temozolomide was documented in a randomized phase III (EORTC-NCIC CE.3) trial, and this therapy, following combined temozolomide and radiation, is the standard of care for patients with newly diagnosed glioblastoma. We comment on the differences in the length of adjuvant therapy in both clinical practice and national studies (e.g. RTOG 0825), usually doubling the length in the EORTC/NCIC study, and relate to historic adjuvant trials for solid tumors.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Dacarbazine/therapeutic use , Glioblastoma/radiotherapy , Humans , Temozolomide , Treatment Outcome
11.
Cancer Chemother Pharmacol ; 69(1): 107-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21614470

ABSTRACT

PURPOSE: Temozolomide (TMZ) is a widely used oral alkylating agent that has been associated with the development of severe hematologic adverse events (HAEs). Limited clinical information about HAEs is available. METHODS: We searched the FDA MedWatch database for TMZ and obtained all MedWatch reports on TMZ submitted to the FDA from November 1, 1997 to September 3, 2008. We defined major HAEs, namely agranulocytosis, aplasia, aplastic anemia (AA), leukemia (various), myelodysplastic syndrome (MDS), and lymphoma, and several minor HAEs. RESULTS: A total of 5,127 reports on 3,490 patients were submitted to MedWatch. Among these, we identified 112 cases of major HAEs. Of the 44 reported deaths, the major HAE was considered the cause in 32 cases. The median duration of TMZ treatment was 6 weeks [0.5-108 weeks]. Seventy-six cases of AA or aplasia and 17 cases of leukemia represented the most common major HAE. Important minor HAEs were bone marrow failure and pancytopenia/pancytopenia-like with 325 combined cases; these reports are clinically similar to aplastic anemia. CONCLUSION: The hematologic toxicity profile of TMZ differs from that of other alkylating agents. TMZ HAEs are emerging as significant concerns. Among alkylating agents, AA appears unique to TMZ, and the high rate warrants disclosure of patients. The duration of TMZ exposure prior to the development of AA may be quite short. The risk of AML/MDS is low, but the length of follow-up is insufficient to assess the true risk.


Subject(s)
Adverse Drug Reaction Reporting Systems/statistics & numerical data , Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/analogs & derivatives , Hematologic Diseases/chemically induced , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Databases, Factual , Humans , Risk , Temozolomide , Time Factors , United States , United States Food and Drug Administration
12.
Cancer Chemother Pharmacol ; 64(4): 647-55, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19543728

ABSTRACT

Temozolomide (TMZ) is an oral alkylating agent that is regarded as a tolerable and effective drug. When combined with radiotherapy in patients with newly diagnosed glioblastoma, survival is significantly prolonged. This finding has led to widespread use of TMZ for patients with this disease. We summarize developing concerns regarding the use of TMZ, imaging of malignant gliomas, and the pharmacology of TMZ-mechanism of action, scheduling and strategies for overcoming resistance.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/adverse effects , Dacarbazine/pharmacokinetics , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Humans , Temozolomide
13.
Am J Health Syst Pharm ; 66(11): 999-1013, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19451611

ABSTRACT

PURPOSE: The adverse events associated with bevacizumab therapy are characterized, and the underlying pathophysiology, risk factors, frequency, and management of these events are described. SUMMARY: The adverse events associated with bevacizumab include hypertension, proteinuria, thromboembolism, impaired wound healing, bleeding, perforation, reversible leukoencephalopathy syndrome, skin rash, and infusion-related hypersensitivity reactions. Patients should be monitored for these events throughout the course of bevacizumab therapy. Hypertension is by far the most common adverse event associated with bevacizumab. Blood pressure should be routinely monitored, and hypertension should be medically managed with antihypertensive drugs as deemed appropriate during bevacizumab therapy. Patients should be monitored for proteinuria every three to four weeks, and bevacizumab should be discontinued with persistent proteinuria of >2+. Thromboembolic events, impaired wound healing, bowel and nasal septum perforation, and bleeding share similar pathophysiology. Thromboembolic events should be managed in accordance with guidelines established by the American College of Chest Physicians, and bevacizumab should be discontinued for new life-threatening venous or arterial thromboembolism. To minimize the risk of bleeding or impaired wound healing, bevacizumab should be started at least four weeks after surgery or discontinued for at least six to eight weeks before elective surgery. The management of other adverse events is more anecdotal, with relatively few reports of their occurrence with bevacizumab. CONCLUSION: Many of the potential serious complications of bevacizumab can be averted by close monitoring of patient-specific variables, which should be measured at baseline and then at predetermined intervals throughout the course of therapy to maximize patient safety.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Bevacizumab , Exanthema/chemically induced , Exanthema/drug therapy , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Humans , Hypertension/chemically induced , Hypertension/drug therapy , Intestinal Perforation/chemically induced , Intestinal Perforation/drug therapy , Posterior Leukoencephalopathy Syndrome/chemically induced , Posterior Leukoencephalopathy Syndrome/drug therapy , Proteinuria/chemically induced , Proteinuria/drug therapy , Thromboembolism/chemically induced , Thromboembolism/drug therapy , Wound Healing/drug effects
14.
Anticancer Drugs ; 18(7): 827-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17581306

ABSTRACT

Cetuximab (Erbitux) is a recombinant, chimeric monoclonal antibody that binds with high affinity to the extracellular ligand-binding domain of human epidermal growth factor receptor. We report a case of repeated responses to cetuximab in a patient with nonresectable squamous cell skin cancer having strong human epidermal growth factor receptor expression. Nonmelanoma skin cancer is the most common cancer in the US, with more than 1 million new cases detected annually, of which 20-25% are squamous cell carcinomas. Although most primary cutaneous squamous cell carcinomas have a high clinical cure rate and are easily treated, the small subset of cancers that recur or metastasize has a poor prognosis, and accounts for approximately 2000 deaths per year. Treatment with cetuximab should be considered for human epidermal growth factor receptor expressing tumors in a palliative setting.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Skin Neoplasms/drug therapy , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Cetuximab , ErbB Receptors , Gene Expression , Humans , Male , Palliative Care , Prognosis , Recurrence
15.
Cancer Immunol Immunother ; 55(9): 1033-42, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16283303

ABSTRACT

T cell immunotherapy of prostate cancer (CaP) offers the potential for less toxic, more effective outcomes. A clinical trial was conducted in 28 patients with locally advanced or metastatic CaP to determine whether an HLA-A2 binding epitope of prostate-specific antigen, PSA146-154 (PSA-peptide), can induce specific T cell immunity. Patients were vaccinated either by intradermal injection of PSA-peptide and GM-CSF or by intravenous administration of autologous dendritic cells pulsed with PSA-peptide at weeks 1, 4 and 10. Delayed-type hypersensitivity (DTH) skin testing was performed at weeks 4, 14, 26 and 52. Fifty percent of the patients developed positive DTH responses to PSA-peptide. The size of the DTH induration progressively increased over time in the majority of responding patients. Skin biopsies from seven DTH-positive patients were available and T cells that developed in situ were also characterized. The phenotype of recovered T cells demonstrated variable proportions of CD4+CD8-, CD4-CD8+ and CD4+CD8+ T cell populations. Cytokine analysis of PSA-peptide stimulated T cells per bead array assay exhibited specific IFN-gamma and TNF-alpha response in six of seven patients. Specific IL-4 response was observed in five patients, while IL-10 response was detected in one patient. Purified CD4-CD8+ T cells isolated from four patients demonstrated specific cytolytic activity per chromium release assay. In conclusion, immunization with PSA-peptide induced specific T cell immunity in one-half of the patients with locally advanced and hormone-sensitive, metastatic CaP. DTH-derived T cells exhibited PSA-peptide-specific cytolytic activity and predominantly expressed a type-1 cytokine profile.


Subject(s)
Cancer Vaccines/immunology , Hypersensitivity, Delayed/immunology , Immunotherapy, Active/methods , Peptide Fragments/immunology , Peptides/immunology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Aged , Aged, 80 and over , Amino Acid Sequence , Cancer Vaccines/administration & dosage , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Epitopes , Humans , Immunity, Cellular , Male , Middle Aged , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptides/administration & dosage , Prostate-Specific Antigen/administration & dosage , Prostatic Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Transplantation, Autologous
16.
Blood ; 99(11): 3905-8, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12010787

ABSTRACT

We have previously demonstrated that 5-aza-2'-deoxycytidine (decitabine) augments fetal hemoglobin (HbF) levels in patients with sickle cell anemia (SS) who did not respond to hydroxyurea (HU). The present study was designed to determine the effect of repeated decitabine dosing on HbF levels and hematologic toxicity over a 9-month treatment period. Seven patients (5 HU nonresponders) were entered. One patient had alpha-thalassemia sickle cell anemia. Decitabine was administered by intravenous infusion at a starting dose of 0.3 mg/kg per day, 5 days a week for 2 weeks, followed by a 4-week observation period. If the absolute neutrophil count dropped below 1000, the dose was reduced by 0.05 mg/kg per day in the next cycle. A drug dose was obtained for each patient, and it resulted in an elevated HbF without neutropenia (absolute neutrophil count nadir greater than 1500) or evidence of cumulative toxicity. Average HbF and average maximal HbF levels attained during the last 20 weeks of treatment for the 6 SS patients increased to 13.93% +/- 2.75% and 18.35% +/- 4.46%, respectively, from a pretreatment mean of 3.12% +/- 2.75%. Mean and mean maximal hemoglobin (Hb) levels increased from 7.23 +/- 2.35 g/dL to 8.81 +/- 0.42 g/dL and 9.73 +/- 0.53 g/dL, respectively. Individual maximal F-cell number observed during the trial was 69% +/- 10.12%. The absence of cumulative toxicity may allow shorter intervals between drug treatments, which may lead to higher hemoglobin and HbF levels after several treatment cycles and, therefore, to greater clinical improvement.


Subject(s)
Anemia, Sickle Cell/drug therapy , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Fetal Hemoglobin/metabolism , Anemia, Sickle Cell/blood , Azacitidine/administration & dosage , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Fetal Hemoglobin/drug effects , Humans , Injections, Subcutaneous , Leukocyte Count , Neutrophils , Reticulocyte Count , Time Factors
17.
Blood ; 102(12): 3865-70, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12907443

ABSTRACT

Fetal hemoglobin (HbF) decreases polymerization of sickle hemoglobin (HbS) and improves outcomes in sickle cell disease (SSD). Therefore, a therapeutic goal in SSD is pharmacologic reactivation of HbF. Silencing of the gamma-globin (HbF) gene is associated with DNA methylation. The cytosine analog 5-aza-2'-deoxycytidine (decitabine) hypomethylates DNA by inhibiting DNA methyltransferase. We examined if subcutaneous decitabine could increase HbF levels and improve SSD pathophysiology without cytotoxicity. Eight symptomatic SSD patients resistant or intolerant of standard treatment with hydroxyurea received decitabine 0.2 mg/kg subcutaneously 1 to 3 times per week in 2 cycles of 6-week duration. Treatment decreased neutrophils and increased mean HbF (6.5% to 20.4%, P <.0001) and mean total hemoglobin (76 to 96 g/L [7.6 to 9.6 g/dL], P <.001). Features of vaso-occlusive crisis pathophysiology such as red cell adhesion, endothelial damage, and coagulation pathway activity significantly improved. gamma-Globin gene promoter methylation decreased, and platelets and the proportion of megakaryocytes and erythroid cells in the marrow increased without a decrease in marrow cellularity, consistent with a DNA hypomethylating, noncytotoxic mechanism of action. Weekly subcutaneous decitabine produces cumulative increases in HbF and total hemoglobin through a noncytotoxic mechanism of action. Chronic dosing and sustained increases in hemoglobin F and total hemoglobin levels may be possible. Further studies in SSD and thalassemia are indicated.


Subject(s)
Anemia, Sickle Cell/drug therapy , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Erythrocytes/drug effects , Fetal Hemoglobin/analysis , Hematopoiesis/drug effects , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/complications , Azacitidine/administration & dosage , Biomarkers/analysis , Blood Coagulation/drug effects , Bone Marrow/pathology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , DNA Methylation/drug effects , Decitabine , Endothelium, Vascular/pathology , Erythrocytes/pathology , Female , Fetal Hemoglobin/drug effects , Humans , Inflammation , Laminin/metabolism , Male , Middle Aged , Thrombospondins/metabolism , Vascular Diseases/etiology , Vascular Diseases/prevention & control
18.
J Oncol Pract ; 4(3): 140-1, 2008 May.
Article in English | MEDLINE | ID: mdl-20856620
SELECTION OF CITATIONS
SEARCH DETAIL