Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Future Oncol ; : 1-12, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38864490

RESUMO

Aims: We assessed care in cancer patients with cachexia across leading health systems (LHSs). Patients & methods: Qualitative interviews and quantitative surveys were conducted with LHSs executives and frontline health care personnel, representing 46 total respondents and 42 unique LHSs and including oncology service line leaders, supportive care services, dietitians and surgical oncologists. Results: Cachexia was not considered a top priority, and formal diagnoses were rare. Participants highlighted the importance of addressing barriers to increase clinical trial enrollment and support frontline health care personnel and patients in early detection of cachexia. Conclusion: Cachexia prioritization needs to be elevated across LHSs executives to obtain capital and strategic imperatives to advance related care.


[Box: see text].

2.
Invest New Drugs ; 38(3): 800-811, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31297636

RESUMO

Background In pancreatic ductal adenocarcinoma (PDAC), the chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-C motif) receptor 2 (CCR2) axis plays a key role in immunosuppressive properties of the tumor microenvironment, patient prognosis, and chemoresistance. This phase Ib study assessed the effects of the orally administered CCR2 inhibitor PF-04136309 in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic PDAC. Methods Patients received PF-04136309 twice daily (BID) continuously plus nab-paclitaxel (125 mg/m2) and gemcitabine (1000 mg/m2) administered on days 1, 8, and 15 of each 28-day cycle. The primary objectives were to evaluate safety and tolerability, characterize dose-limiting toxicities (DLTs), and determine the recommended phase II dose (RP2D) of PF-04136309. Results In all, 21 patients received PF-04136309 at a starting dose of 500 mg or 750 mg BID. The RP2D was identified to be 500 mg BID. Of 17 patients treated at the 500 mg BID starting dose, three (17.6%) experienced a total of four DLTs, including grade 3 dysesthesia, diarrhea, and hypokalemia and one event of grade 4 hypoxia. Relative to the small number of patients (n = 21), a high incidence (24%) of pulmonary toxicity was observed in this study. The objective response rate for 21 patients was 23.8% (95% confidence interval: 8.2-47.2%). Levels of CD14 + CCR2+ inflammatory monocytes (IM) decreased in the peripheral blood, but did not accumulate in the bone marrow. Conclusions PF-04136309 in combination with nab-paclitaxel plus gemcitabine had a safety profile that raises concern for synergistic pulmonary toxicity and did not show an efficacy signal above nab-paclitaxel and gemcitabine. ClinicalTrials.gov identifier: NCT02732938.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Quimiocina CCL2/antagonistas & inibidores , Neoplasias Pancreáticas/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/uso terapêutico , Adenocarcinoma/metabolismo , Idoso , Albuminas/uso terapêutico , Carcinoma Ductal Pancreático/metabolismo , Estudos de Coortes , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/metabolismo , Prognóstico , Pirrolidinas/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Gencitabina , Neoplasias Pancreáticas
3.
Future Oncol ; 15(28): 3267-3281, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31394933

RESUMO

More oncology biologics are becoming available for subcutaneous (sc.) administration and are expected to provide useful therapeutic options. We evaluated evidence published in the past 5 years to assess the humanistic and economic impact of sc. versus intravenous administration of approved cancer therapies and identify outcomes favoring either administration route. These publications focused predominantly on healthcare resource utilization and economic outcomes, demonstrating resource and cost savings with sc. administration. Patients reported a better health-related quality of life and preference for sc. formulations. Time-and-motion study analyses confirmed the convenience of sc. administration. These findings suggest that future availability of sc. oncology biologics, especially anti-PD-1/PD-ligand 1 antibodies due to their increased utility in various malignancies, may be beneficial for patients, healthcare providers and payers.


Assuntos
Antineoplásicos/economia , Produtos Biológicos/economia , Análise Custo-Benefício , Neoplasias/tratamento farmacológico , Neoplasias/economia , Administração Intravenosa , Antineoplásicos/administração & dosagem , Produtos Biológicos/administração & dosagem , Humanos , Injeções Subcutâneas , Neoplasias/patologia , Prognóstico
4.
Cancer Biol Ther ; 22(2): 89-105, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33427569

RESUMO

Recently approved therapies have contributed to a significant progress in the management of ovarian cancer; yet, more options are needed to further improve outcomes in patients with advanced disease. Here we review the rationale and ongoing clinical trials of novel combination strategies involving chemotherapy, poly ADP ribose polymerase, programmed death 1 (PD-1)/PD-ligand 1 immune checkpoint and/or vascular endothelial growth factor receptor inhibitors. Further, we discuss novel agents aimed at targets associated with ovarian cancer growth or progression that are emerging as potential new treatment approaches. Among them, agents targeted to folate receptor α, tissue factor, and protein kinase-mediated pathways (WEE1 kinase, phosphatidylinositol-3 kinase α, cell cycle checkpoint kinase 1/2, ATR kinase) are currently in clinical development as mono- or combination therapies. If successful, findings from these extensive development efforts may further transform treatment of patients with advanced ovarian cancer.


Assuntos
Carcinoma Epitelial do Ovário/tratamento farmacológico , Inibidores de Checkpoint Imunológico/uso terapêutico , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacologia
5.
Paediatr Drugs ; 23(4): 381-394, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34173206

RESUMO

Regulatory changes have been enacted in the United States (US) and European Union (EU) to encourage the development of new treatments for pediatric cancer. Here, we review some of the factors that have hampered the development of pediatric cancer treatments and provide a comparison of the US and EU regulations implemented to address this clinical need. We then provide some recommendations for each stage of the oncology drug development pathway to help researchers maximize their chance of successful drug development while complying with regulations. A key recommendation is the engagement of key stakeholders such as regulatory authorities, pediatric oncologists, academic researchers, patient advocacy groups, and a Pediatric Expert Group early in the drug development process. During drug target selection, sponsors are encouraged to consult the Food and Drug Administration (FDA), European Medicines Agency (EMA), and the FDA target list, in addition to relevant US and European consortia that have been established to characterize and prioritize oncology drug targets. Sponsors also need to carefully consider the resourcing requirements for preclinical testing, which include ensuring appropriate access to the most relevant databases, clinical samples, and preclinical models (cell lines and animal models). During clinical development, sponsors can account for the pharmacodynamic (PD)/pharmacokinetic (PK) considerations specific to a pediatric population by developing pediatric formulations, selecting suitable PD endpoints, and employing sparse PK sampling or modeling/simulation of drug exposures where appropriate. Additional clinical considerations include the specific design of the clinical trial, the potential inclusion of children in adult trials, and the value of cooperative group trials.


In the last few decades, great progress has been made in developing new treatments for adult cancers. However, development of new treatments for childhood cancers has been much slower. To encourage drug companies (sponsors) to develop effective treatments for childhood cancer, authorities in the United States (US) and Europe have made new rules for drug development. Under these new rules, sponsors developing drugs for specific cancers in adults have to consider whether the target of that drug also causes cancers in children. If this is the case, sponsors have to carry out clinical studies of their drug in children who have cancer that is caused by the same drug target. In this article, we describe some reasons for why drug development for childhood cancers has been slow and the rules created to address this problem in the US and Europe. We share some recommendations to help sponsors maximize their chances of developing an effective drug in children while satisfying the new rules. Specifically, sponsors need to be aware of the differences between studying drugs in adults versus children and how these influence the way the drug is tested. We make several recommendations for each stage of the development process, beginning with what is needed even before human studies begin. Finally, we highlight some issues that sponsors need to think about during drug development, from the preclinical stage (testing drugs in cells and animals) through to clinical testing in adults and pediatric patients with cancer.


Assuntos
Antineoplásicos/uso terapêutico , Desenvolvimento de Medicamentos/legislação & jurisprudência , Prova Pericial/legislação & jurisprudência , Oncologia/legislação & jurisprudência , Neoplasias/tratamento farmacológico , Antineoplásicos/síntese química , Criança , Ensaios Clínicos como Assunto/legislação & jurisprudência , Ensaios Clínicos como Assunto/métodos , Sistemas de Liberação de Medicamentos/métodos , Desenvolvimento de Medicamentos/métodos , União Europeia , Prova Pericial/métodos , Humanos , Oncologia/métodos , Neoplasias/epidemiologia , Estados Unidos , United States Food and Drug Administration/legislação & jurisprudência
6.
Target Oncol ; 16(6): 773-787, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34694529

RESUMO

BACKGROUND: Sasanlimab is a monoclonal antibody that binds to the programmed cell death receptor 1 (PD-1). Anti-PD-1 monoclonal antibodies have improved patient clinical outcomes; however, not all treated patients derive clinical benefit. Further insights on potential biomarkers beyond PD-L1 expression levels would help to identify the patients most likely to respond to treatment. OBJECTIVE: This study evaluated tumor biopsies from patients treated with intravenous or subcutaneous sasanlimab to identify biomarkers of response and characterize pharmacodynamic activity. METHODS: Anti-PD-1/PD-ligand 1 (PD-L1)-naive patients with advanced solid tumors received sasanlimab intravenously at 1, 3, or 10 mg/kg every 3 weeks (n = 23) or subcutaneously at 300 mg every 4 weeks (n = 15). Best tumor percentage change from baseline was determined by RECIST. Whole-exome DNA and RNA sequencing were performed in tumor samples collected from treated patients at protocol-defined timepoints. PD-L1 and CD8 protein expression were evaluated in tumor biopsies by immunohistochemistry. Associations with response were assessed by linear regression analysis. RESULTS: Baseline tumor mutational burden (TMB), as well as PD-L1 and CD8 expression, were significantly associated with response to sasanlimab across the multiple dose levels, routes of administration, and range of tumor types evaluated. TMB is an independent biomarker from the various tumor inflammatory genes and signatures evaluated. Gene set enrichment analysis showed that higher baseline expression levels of genes related to the interferon-γ and PD-1 signaling pathways and the cell cycle were significantly associated with response to sasanlimab across tumor types. No differences were observed between routes of administration with regard to response to sasanlimab for the biomarkers of interest (TMB, PD-L1, CD8, and interferon-γ signature). Evaluation of pharmacodynamic changes showed increased tumor expression of genes enriched in adaptive immune response pathways. CONCLUSIONS: Our findings indicate an active, immunomodulatory mechanism for the anti-PD-1 antibody sasanlimab across different tumor types and routes of administration. TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT02573259; registered October 2015.


Assuntos
Antígeno B7-H1 , Neoplasias , Anticorpos Monoclonais/uso terapêutico , Biomarcadores Tumorais/metabolismo , Expressão Gênica , Humanos , Inibidores de Checkpoint Imunológico , Interferon gama/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Receptor de Morte Celular Programada 1
7.
Clinicoecon Outcomes Res ; 12: 693-709, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262624

RESUMO

PURPOSE: Non-muscle invasive bladder cancer (NMIBC) is a malignancy restricted to the inner lining of the bladder. Intravesical Bacillus Calmette-Guerin (BCG) following transurethral resection of the bladder tumor is the mainstay first-line treatment for high-risk NMIBC patients. Two systematic literature reviews (SLRs) were conducted to further assess the current evidence on BCG use in NMIBC and the humanistic and economic burden of disease. METHODS: Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, Embase® and MEDLINE® were searched using the Ovid platform to identify interventional or real-world evidence studies on the health-related quality of life (HRQoL) and economic burden in NMIBC. Limited evidence was found from initial economic SLR searches in NMIBC, so additional targeted searches for bladder cancer were conducted to expand findings. RESULTS: Fifty-nine publications were included in the HRQoL SLR, of which 23 reported HRQoL and symptoms in NMIBC. At diagnosis, HRQoL was comparable with population norms but worsened considerably 2 years following diagnosis. Maintenance therapy with intravesical BCG was associated with reduced HRQoL, and treatment-related adverse events (AEs) resembled typical NMIBC symptoms. Twenty-two studies reported decreasing BCG compliance over time. Common AEs with BCG were frequent urination, lower urinary tract symptoms, pain, and hematuria. Forty-two publications were included in the economic SLR, of which nine assessed healthcare costs and resource use in NMIBC or bladder cancer. High-risk disease and high-intensity treatment were associated with increased healthcare costs. CONCLUSION: NMIBC has a considerable symptomatic, HRQoL, and economic burden. Symptoms persisted and HRQoL worsened despite intravesical BCG treatment. NMIBC is a costly disease, with higher healthcare costs associated with increased risk of disease progression and recurrence. There is a high unmet need for safe and effective treatments that reduce the risk of disease progression and recurrence, provide symptomatic relief, and improve HRQoL for patients.

8.
Cancer Treat Rev ; 76: 1-9, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30913454

RESUMO

Although recent advances in the treatment of castration-resistant prostate cancer (CRPC) have significantly improved patient outcomes, advanced prostate cancer is still associated with substantial morbidity and mortality, particularly in patients who develop resistance after multiple lines of therapy. Various cell signaling, DNA repair, and epigenetic enzymatic pathways are being targeted with small-molecule inhibitors in order to identify treatment strategies for patients with CRPC. In this review, we discuss novel targets and agents, studied preclinically and now being validated in clinical trials, including poly ADP-ribose polymerase (PARP), enhancer of zeste homologue 2 (EZH2), hedgehog pathway, MDM2/p53, and tyrosine kinase inhibitors. Further, we outline current approaches for novel prostate cancer vaccines such as DCVAC/PCa, PROSTVAC-V/F, MVI-816, CV9104, and PF-06753512. This wide spectrum of potential treatment strategies holds promise for additional improvements in the treatment of patients with CRPC, as these novel agents are aimed at targets known to be associated with growth and malignant progression of prostate cancer. If primary study endpoints are met, findings from ongoing phase III trials of well-tolerated and active combinations may provide new effective treatment options for advanced prostate cancer and thereby contribute to enhanced disease control in CRPC patients.


Assuntos
Neoplasias de Próstata Resistentes à Castração/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Humanos , Masculino , Terapia de Alvo Molecular/métodos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/imunologia , Ensaios Clínicos Controlados Aleatórios como Assunto
9.
J Immunother Cancer ; 7(1): 105, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30992085

RESUMO

The increasing use of multiple immunomodulatory (IMD) agents for cancer therapies (e.g. antibodies targeting immune checkpoints, bispecific antibodies, and chimeric antigen receptor [CAR]-T cells), is raising questions on their potential immunogenicity and effects on treatment. In this review, we outline the mechanisms of action (MOA) of approved, antibody-based IMD agents, potentially related to their immunogenicity, and discuss the reported incidence of anti-drug antibodies (ADA) as well as their clinical relevance in patients with cancer. In addition, we discuss the impact of the administration route and potential strategies to reduce the incidence of ADA and manage treated patients. Analysis of published reports indicated that the risk of immunogenicity did not appear to correlate with the MOA of anti-programmed death 1 (PD-1)/PD-ligand 1 monoclonal antibodies nor to substantially affect treatment with most of these agents in the majority of patients evaluated to date. Treatment with B-cell depleting agents appears associated with a low risk of immunogenicity. No significant difference in ADA incidence was found between the intravenous and subcutaneous administration routes for a panel of non-oncology IMD antibodies. Additionally, while the data suggest a higher likelihood of immunogenicity for antibodies with T-cell or antigen-presenting cell (APC) targets versus B-cell targets, it is possible to have targets expressed on APCs or T cells and still have a low incidence of immunogenicity.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Doenças do Sistema Imunitário/imunologia , Imunoterapia Adotiva/efeitos adversos , Depleção Linfocítica/efeitos adversos , Neoplasias/terapia , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/imunologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Humanos , Doenças do Sistema Imunitário/induzido quimicamente , Doenças do Sistema Imunitário/epidemiologia , Doenças do Sistema Imunitário/prevenção & controle , Imunossupressores/uso terapêutico , Imunoterapia Adotiva/métodos , Incidência , Depleção Linfocítica/métodos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptores de Antígenos Quiméricos/imunologia , Resultado do Tratamento
10.
Target Oncol ; 12(5): 599-610, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28801849

RESUMO

Colorectal cancer (CRC) is a leading cause of tumor-related morbidity and mortality worldwide, with mortality most often attributable to metastatic disease. Bevacizumab, a humanized monoclonal antibody targeting vascular endothelial growth factor, has a significant role in the treatment of metastatic CRC (mCRC). However, patient access to bevacizumab may be limited in some regions or circumstances, owing to factors related to insurance coverage, reimbursement, patient out-of-pocket costs, or availability. As a result, outcomes for patients with mCRC may be worsened. Additionally, counterfeit bevacizumab has infiltrated legitimate supply chains, exposing patients to risk. Oncologists may also be affected detrimentally, since resolving access issues can be time-consuming and demoralizing. The imminent expiry of patents protecting bevacizumab provides other manufacturers with the opportunity to produce highly similar versions known as biosimilars. High-quality, safe, and effective biosimilars have the potential to expand access to bevacizumab. Most of the bevacizumab biosimilars currently in development are in clinical trials in patients with non-small-cell lung cancer, and future authorization for mCRC indications will, therefore, be based on extrapolation. This article reviews the current role of bevacizumab in the management of mCRC, the possible barriers associated with diminished access to bevacizumab, and the potential bevacizumab biosimilars in development. How biosimilars may impact the treatment of mCRC is also discussed.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Bevacizumab/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Humanos
11.
J Geriatr Oncol ; 7(1 Suppl): S1-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26856525

RESUMO

Biosimilar monoclonal antibodies are being developed globally to meet clinical demand in oncology and potentially provide greater access to biologic therapies for patients with cancer, including older patients. In this supplement, we present an overview of the development, approval requirements, and characteristics of biosimilar monoclonal antibodies that may help practicing oncologists and other healthcare providers to acquire familiarity with this new group of therapeutic biologic agents. Furthermore, we review and discuss some of the challenges and potential strategies for the management of older patients with cancer, who represent an increasing population in many countries.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Neoplasias/tratamento farmacológico , Idoso , Terapia Biológica , Humanos
12.
Clin Lymphoma Myeloma Leuk ; 16(4): 175-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26906106

RESUMO

Diffuse large B-cell lymphoma (DLBCL), an aggressive non-Hodgkin lymphoma (NHL), is the most-common subtype of NHL. DLBCL can be classified into at least 3 major immunologically distinct types, which contributes to considerable variation in disease prognosis and response to treatment. DLBCL potentially is curable, even when diagnosed at advanced stages. The current standard of care for most patients with untreated or relapsed/refractory DLBCL is chemoimmunotherapy containing rituximab, an anti-CD20 monoclonal antibody. With advanced understanding of the molecular mechanisms involved in the pathogenesis of DLBCL and specific signaling pathways that are activated in different subtypes, potential new therapeutic targets have been identified, some of which are at the late stages of clinical development. This review summarizes the critical role of rituximab in the current standard of care treatment for DLBCL and discusses why rituximab is likely to remain an important component of treatment options for DLBCL in the foreseeable future. In addition, current and emerging therapeutic agents, including potential benefits of rituximab biosimilars, for patients with DLBCL are discussed. The advent of rituximab biosimilars may facilitate accessibility of rituximab-based chemotherapies to patients with DLBCL and has potential cost-saving benefits for healthcare systems globally.


Assuntos
Antineoplásicos/uso terapêutico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Rituximab/uso terapêutico , Adulto , Idoso , Antineoplásicos/farmacologia , Medicamentos Biossimilares , Humanos , Imunoterapia/métodos , Linfoma Difuso de Grandes Células B/patologia , Pessoa de Meia-Idade , Prognóstico , Rituximab/farmacologia
13.
BioDrugs ; 30(3): 195-206, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27097915

RESUMO

Biopharmaceuticals have the potential to raise an immunogenic response in treated individuals, which may impact the efficacy and safety profile of these drugs. As a result, it is essential to evaluate immunogenicity throughout the different phases of the clinical development of a biopharmaceutical, including post-marketing surveillance. Although rigorous evaluation of biopharmaceutical immunogenicity is required by regulatory authorities, there is a lack of uniform standards for the type, quantity, and quality of evidence, and for guidance on experimental design for immunogenicity assays or criteria to compare immunogenicity of biopharmaceuticals. Moreover, substantial technological advances in methods to assess immune responses have yielded higher immunogenicity rates with modern assays, and limit comparison of immunogenicity of biopharmaceuticals outside of head-to-head clinical trials. Accordingly, research programs, regulatory agencies, and clinicians need to keep pace with continuously evolving analyses of immunogenicity. Here, we review factors associated with immunogenicity of biopharmaceuticals, potential clinical ramifications, and current regulatory guidance for evaluating immunogenicity, and discuss methods to assess immunogenicity in non-clinical and clinical studies. We also describe special considerations for evaluating the immunogenicity of biosimilar candidates.


Assuntos
Produtos Biológicos/imunologia , Biofarmácia/normas , Adalimumab/imunologia , Anticorpos/análise , Produtos Biológicos/efeitos adversos , Produtos Biológicos/farmacologia , Biofarmácia/métodos , Medicamentos Biossimilares/farmacologia , Humanos , Estados Unidos , United States Food and Drug Administration
14.
MAbs ; 8(5): 861-6, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27050730

RESUMO

Biosimilars are designed to be highly similar to approved or licensed (reference) biologics and are evaluated based on the totality of evidence from extensive analytical, nonclinical and clinical studies. As part of the stepwise approach recommended by regulatory agencies, the first step in the clinical evaluation of biosimilarity is to conduct a pharmacokinetics similarity study in which the potential biosimilar is compared with the reference product. In the context of biosimilar development, a pharmacokinetics similarity study is not necessarily designed for a comparative assessment of safety. Development of PF-05280014, a potential biosimilar to trastuzumab, illustrates how a numerical imbalance in an adverse event in a small pharmacokinetics study can raise questions on safety that may require additional clinical trials.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Medicamentos Biossimilares/efeitos adversos , Aprovação de Drogas , Ensaios Clínicos Fase I como Assunto , Método Duplo-Cego , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
15.
MAbs ; 7(4): 653-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25961747

RESUMO

Biosimilar monoclonal antibodies are being developed globally for patients with different types of solid tumors and hematologic malignancies. Applications for proposed biosimilar monoclonal antibodies are being submitted to the regulatory authorities around the world and may increase patient access to key treatment options upon approval. An understanding among stakeholders (e.g., physicians, patients and their caregivers, pharmacists, payers) of the approval criteria, as well as the similarities and differences in regulatory pathways involved in biosimilar approval in different countries, as presented in this review, will facilitate identification of high-quality, safe, monoclonal antibodies that have been developed according to strict, biosimilar regulatory standards. Further guidance and resolution of the ongoing discussions on biosimilar labeling, naming, automatic substitution, and indication extrapolation may ensure, in the future, an effective and appropriate use of biosimilar monoclonal antibodies by oncologists and other stakeholders in daily clinical practice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Medicamentos Biossimilares , Aprovação de Drogas , Legislação de Medicamentos , Humanos
16.
Drug Discov Today ; 20 Suppl 2: 1-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25572407

RESUMO

The widespread use and patent expiration of many biologics have led to global interest in development of biosimilar products. Because the manufacture of biologics, including biosimilars, is a complex process involving living systems, the development of a biosimilar is more rigorous than the development of a generic small molecule drug. Several regulatory agencies have established or are proposing guidelines that recommend a stepwise process to ensure the efficacy and safety of a biosimilar are highly similar to the reference product. This article also explores the early clinical phase of biosimilar development, which is particularly important to resolving any uncertainties that might remain following in vitro and in vivo evaluations and to enable a selective and targeted approach to Phase III clinical efficacy and safety investigation.


Assuntos
Medicamentos Biossimilares/farmacologia , Aprovação de Drogas , Descoberta de Drogas/métodos , Indústria Farmacêutica/métodos , United States Food and Drug Administration , Animais , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/farmacocinética , Medicamentos Biossimilares/normas , Ensaios Clínicos como Assunto , Qualidade de Produtos para o Consumidor , Aprovação de Drogas/legislação & jurisprudência , Descoberta de Drogas/legislação & jurisprudência , Descoberta de Drogas/normas , Avaliação Pré-Clínica de Medicamentos , Indústria Farmacêutica/legislação & jurisprudência , Indústria Farmacêutica/normas , Europa (Continente) , Guias como Assunto , Humanos , Controle de Qualidade , Medição de Risco , Estados Unidos , United States Food and Drug Administration/legislação & jurisprudência , United States Food and Drug Administration/normas , Organização Mundial da Saúde
17.
Drug Des Devel Ther ; 9: 3247-55, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26150698

RESUMO

Acute and chronic shortages of various pharmaceuticals and particularly of sterile injectable products are being reported on a global scale, prompting evaluation of more effective strategies to manage current shortages and development of new, high-quality pharmaceutical products to mitigate the risk of potential future shortages. Oncology drugs such as liposomal doxorubicin and 5-fluorouracil represent examples of first-choice drugs critically affected by shortages. Survey results indicate that the majority of hospitals and practicing oncologists have experienced drug shortages, which may have compromised patient safety and clinical outcomes, and increased health care costs, due to delays or changes in treatment regimens. Clinical trials evaluating novel agents in combination with standard-of-care drugs are also being affected by drug shortages. Clinical and ethical considerations on treatment objectives, drug indication, and availability of alternative options may help in prioritizing cancer patients involved in active drug shortages. The United States Food and Drug Administration and the European Medicines Agency have identified manufacturing problems, delays in supply, and lack of available active ingredients as the most frequent causes of recent or ongoing drug shortages, and have released specific guidance to monitor, manage, and reduce the risk of shortages. The upcoming loss of exclusivity for a number of anticancer biologics, together with the introduction of an abbreviated approval pathway for biosimilars, raises the question of whether these products will be vulnerable to shortages. Future supply by reliable manufacturers of well characterized biosimilar monoclonal antibodies, developed in compliance with regulatory and manufacturing guidelines and with substantial investments, may contribute to prevent future biologics shortages and ensure access to effective and safe treatment options for patients with cancer. Preclinical and clinical characterization is ongoing for potential biosimilars of trastuzumab, rituximab, and bevacizumab, with promising results.


Assuntos
Antineoplásicos/provisão & distribuição , Medicamentos Biossimilares/provisão & distribuição , Atenção à Saúde , Indústria Farmacêutica , Neoplasias/tratamento farmacológico , Antineoplásicos/efeitos adversos , Antineoplásicos/economia , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/economia , Atenção à Saúde/economia , Atenção à Saúde/tendências , Custos de Medicamentos , Indústria Farmacêutica/economia , Indústria Farmacêutica/tendências , Previsões , Humanos , Neoplasias/economia , Segurança do Paciente , Medição de Risco , Fatores de Risco
18.
Drug Discov Today ; 20 Suppl 1: 3-15, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25912284

RESUMO

Biosimilar development requires several steps: selection of an appropriate reference biologic, understanding the key molecular attributes of that reference biologic and development of a manufacturing process to match these attributes of the reference biologic product. The European Medicines Agency (EMA) and the FDA guidance documents state that, in lieu of conducting extensive preclinical and clinical studies typically required for approval of novel biologics, biosimilars must undergo a rigorous similarity evaluation. The aim of this article is to increase understanding of the preclinical development and evaluation process for biosimilars, as required by the regulatory agencies, that precedes the clinical testing of biosimilars in humans.


Assuntos
Medicamentos Biossimilares/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica/métodos , Animais , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/farmacocinética , Medicamentos Biossimilares/normas , Qualidade de Produtos para o Consumidor , Avaliação Pré-Clínica de Medicamentos/normas , Indústria Farmacêutica/normas , Humanos , Estrutura Molecular , Controle de Qualidade , Fatores de Risco , Relação Estrutura-Atividade , Equivalência Terapêutica
19.
Arch Surg ; 138(1): 63-6; discussion 67, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12511153

RESUMO

BACKGROUND: Radioisotope mapping is an essential technical component of sentinel lymph node (SLN) biopsy, and most authors define success by an arbitrary threshold SLN-background ratio. HYPOTHESIS: Few studies have examined the degree to which the relative level of SLN counts correlates with the presence of metastasis. Having removed the SLN with the highest counts, there are no data suggesting how far the surgeon should persist in removing additional SLNs that contain much lower levels of isotope. METHODS: We performed 134 SLN biopsy procedures in 132 patients with melanoma. Successful isotope localization was defined using an SLN/"hottest" SLN ratio; we defined an SLN as any node containing counts at least 10% of that of the hottest SLN. RESULTS: Of 83 patients with more than 1 SLN site identified, 21 (25%) had SLNs that contained metastasis. In 17 (81%) of these cases, the SLN with the highest countcontained tumor, but in 4 (19%) it was benign. Among these 4 patients, the counts of the hottest benign SLNs exceeded those of SLNs positive for metastasis on histological examination by a ratio of at least 10:1, and the counts of the positive SLNs were less than 4:1 of those of the background counts or the presence of blue dye failed to identify the positive SLN. No optimum ratio of SLN/SLN or SLN/background counts identified the positive SLN in all cases. CONCLUSIONS: Although the SLN with the highest counts contained metastasis in 81% of patients with malignant melanoma and multiple SLNs, neither a relatively high isotope count nor the presence of blue dye consistently predicted SLN positivity. For maximum accuracy, SLN biopsy requires the removal of all nodes containing isotope regardless of the relative magnitude of counts and the concurrent use of blue dye to salvage those procedures in which isotope mapping fails.


Assuntos
Metástase Linfática/diagnóstico por imagem , Melanoma/patologia , Melanoma/cirurgia , Compostos Radiofarmacêuticos , Biópsia de Linfonodo Sentinela/métodos , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/cirurgia , Coloide de Enxofre Marcado com Tecnécio Tc 99m , Adulto , Idoso , Corantes , Feminino , Humanos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Estudos Prospectivos , Cintilografia , Biópsia de Linfonodo Sentinela/normas
20.
Arch Surg ; 138(10): 1135-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14557133

RESUMO

BACKGROUND: The incidence of cutaneous melanoma is rising steadily, and the rate of increase is among the highest for any form of cancer. Although the reliability of age as a prognostic factor is debatable, several studies suggest that age has an important prognostic use. HYPOTHESIS: Age alone does not predict a poor prognosis in the older patient with melanoma. SETTING: University teaching hospital. METHODS: A retrospective review was undertaken to identify patients aged 65 years or older with intermediate-thickness melanoma (1-4 mm). Two hundred thirteen such patients were identified. Data are given as mean +/- SD. RESULTS: The mean age was 72.2+/-6.1 years. The mean follow-up was 49 months. By univariate analysis, the mean disease-free survival (DFS) and overall survival (OS) for lymph node-positive patients was 36.0+/-9.6 and 56.0+/-10.6 months, respectively. The mean DFS for node-negative patients was 155.0+/-9.8 months, and the mean OS was 166.0+/-9.2 months (P<.001 for both). The mean DFS and OS for women were 151.0+/-11.2 and 163.0+/-10.9 months, respectively. In contrast, men had 116.0+/-9.5 months' DFS and 127.0+/-9.0 months' OS (P=.01 for both). By multivariate analysis, lymph node status was the most predictive variable for DFS and OS (P<.001 for both). Sex tended to affect OS (P=.02) but did not achieve prognostic significance on DFS (P=.09). Other factors such as location, ulceration, histological type, and mitoses per square millimeter failed to show any prognostic significance. Stratification into 3 age groups (65-70, 71-80, and >80 years) had no significant effect on DFS (P=.95) or OS (P=.92). CONCLUSIONS: Lymph node status is the most important prognostic factor in older patients with intermediate-thickness melanoma. Identification of high-risk factors may help stratify these patients for recommendation of more aggressive treatment or adjuvant therapies. Among these patients, age alone was not a significant prognostic factor in the clinical management of melanoma.


Assuntos
Melanoma/epidemiologia , Neoplasias Cutâneas/epidemiologia , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Incidência , Masculino , Melanoma/cirurgia , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Neoplasias Cutâneas/cirurgia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA