Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Curr Oncol Rep ; 26(5): 538-550, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581469

RESUMEN

PURPOSE OF REVIEW: This paper aims to address the latest findings in neuroendocrine tumor (NET) theranostics, focusing on new evidence and future directions of combined diagnosis with positron emission tomography (PET) and treatment with peptide receptor radionuclide therapy (PRRT). RECENT FINDINGS: Following NETTER-1 trial, PRRT with [177Lu]Lu-DOTATATE was approved by FDA and EMA and is routinely employed in advanced G1 and G2 SST (somatostatin receptor)-expressing NET. Different approaches have been proposed so far to improve the PRRT therapeutic index, encompassing re-treatment protocols, combinations with other therapies and novel indications. Molecular imaging holds a potential added value in characterizing disease biology and heterogeneity using different radiopharmaceuticals (e.g., SST and FDG) and may provide predictive and prognostic parameters. Response assessment criteria are still an unmet need and new theranostic pairs showed preliminary encouraging results. PRRT for NET has become a paradigm of modern theranostics. PRRT holds a favorable toxicity profile, and it is associated with a prolonged time to progression, reduction of symptoms, and improved patients' quality of life. In light of further optimization, different new strategies have been investigated, along with the development of new radiopharmaceuticals.


Asunto(s)
Tumores Neuroendocrinos , Octreótido/análogos & derivados , Compuestos Organometálicos , Radiofármacos , Humanos , Tumores Neuroendocrinos/radioterapia , Tumores Neuroendocrinos/diagnóstico por imagen , Radiofármacos/uso terapéutico , Octreótido/uso terapéutico , Tomografía de Emisión de Positrones/métodos , Receptores de Péptidos/uso terapéutico , Receptores de Péptidos/metabolismo , Nanomedicina Teranóstica/métodos , Radioisótopos/uso terapéutico
2.
Eur J Nucl Med Mol Imaging ; 51(4): 1136-1146, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38040931

RESUMEN

PURPOSE: Currently, the most used peptide receptor radionuclide therapy (PRRT) regimen for neuroendocrine tumors comprises 4 treatment cycles, and there is not enough large-scale data to support the safety of more individualized extended PRRT. This study aims to evaluate the therapeutic effectiveness and potential nephrotoxicity related to PRRT using more than four treatment cycles. METHODS: In this retrospective analysis, we included patients who had received at least four PRRT cycles and had available follow-up data. We analyzed renal function indicators before and after multiple treatments, comparing nephrotoxicity in patients receiving four cycles ("standard") with those receiving more than four ("extended treatment"). Nephrotoxicity was assessed via creatinine levels and CTCAE creatinine grades. Treatment effectiveness was gauged using Kaplan-Meier survival analysis, focusing on overall survival and disease-specific survival (DSS). Statistical analyses were performed using SPSS version 26 (IBM), R 4.2.3, and GraphPad Prism 9.0.0. Statistical significance was defined as a P-value of less than 0.05. RESULTS: Our study cohort consisted of 281 patients in the standard group and 356 in the extended treatment group. No significant differences in baseline characteristics or renal function were noted between the two groups pre-treatment. Mean post-treatment creatinine levels did not significantly differ between the standard (89.30 ± 51.19 µmol/L) and extended treatment groups (93.20 ± 55.98 µmol/L; P = 0.364). Similarly, there was no statistical significance between the CTCAE creatinine grades of the two groups (P = 0.448). Adverse renal events were observed in 0.4% of patients in the standard group and 1.1% in the extended treatment group. After a median follow-up time of 88.3 months, we found that median overall survival was significantly higher in the extended treatment group (72.8 months) compared to the standard treatment group (52.8 months). A Cox regression analysis further supported these findings, indicating a better prognosis for the extended treatment group in terms of overall survival (HR: 0.580, P < 0.001) and DSS (HR: 0.599, P < 0.001). CONCLUSION: Our findings suggest that extending PRRT treatment beyond the standard four cycles may be a safe and effective therapeutic strategy for NET patients. This approach could be particularly beneficial for patients experiencing disease recurrence or progression following standard treatment.


Asunto(s)
Tumores Neuroendocrinos , Humanos , Tumores Neuroendocrinos/patología , Estudios Retrospectivos , Creatinina , Octreótido/efectos adversos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Radioisótopos , Receptores de Péptidos/uso terapéutico
3.
Clin Nucl Med ; 48(6): 547-548, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-36928302

RESUMEN

ABSTRACT: We report on a patient diagnosed with an aggressive pancreatic neuroendocrine tumor (NET G3; Ki67 = 60%), who underwent pancreatic resection with partial removal of liver lesions. The patient refused chemotherapy. Dual-tracer imaging with 18 F-FDG and somatostatin receptor (SSTR)-targeted PET/CT was conducted. Radiotracer accumulation on both imaging modalities in bilobar hepatic lesions was observed. "Cold" somatostatin analogues with four cycles of peptide receptor radionuclide therapy (PRRT) were initiated, leading to partial response. Even in highly proliferative but differentiated G3 NET (Ki67>55%), SSTR expression in sites of disease should be evaluated, which may then allow PRRT, even as first-line systemic treatment.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Pancreáticas , Receptores de Péptidos , Receptores de Somatostatina , Humanos , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/terapia , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/terapia , Receptores de Péptidos/metabolismo , Receptores de Péptidos/uso terapéutico , Trazadores Radiactivos , Receptores de Somatostatina/metabolismo , Receptores de Somatostatina/uso terapéutico , Tomografía Computarizada por Tomografía de Emisión de Positrones
4.
Clin Nucl Med ; 48(3): 221-227, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36723881

RESUMEN

PURPOSE: Progressive metastatic medullary thyroid carcinoma (MTC) is often characterized by rapid disease progression and poor prognosis, with only few therapeutic options available. Peptide receptor radionuclide therapy (PRRT) has demonstrated remarkable success in the management of gastroenteropancreatic neuroendocrine tumors and has also been suggested to treat MTC. However, evidence on its effectiveness and long-term outcome for this indication is still limited. The objective of this study was to assess the safety and efficacy of PRRT in patients with advanced, progressive MTC and to determine survival. Potential predictors of survival were also evaluated. METHODS: From September 2003 to June 2019, 28 patients (15 men and 13 women; mean age, 49 ± 14 years) with progressive, somatostatin receptor-positive advanced MTC received PRRT with 177Lu- or 90Y-labeled somatostatin analogs at Zentralklinik Bad Berka, Germany. Toxicity was graded according to Common Terminology Criteria for Adverse Events version 5.0. Treatment response was evaluated according to RECIST (Response Evaluation Criteria in Solid Tumors) 1.1, as well as molecular imaging criteria (European Organisation for Research and Treatment of Cancer). Kaplan-Meier analysis was used to calculate progression-free survival (PFS) and overall survival (OS), defined from the start of PRRT. Univariate and multivariate Cox regression analyses were performed to identify parameters associated with PFS and OS. RESULTS: Seventy-seven cycles of PRRT were administered (mean cumulative administered activity, 16.0 ± 7.8 GBq). No acute or long-term grade 3/4 toxicity was recorded with a follow-up of 3 to 140 months, except for 1 patient (4%) who suffered from grade 3 anemia (possibly related to disease progression). According to the RECIST criteria, the disease control rate after 3 to 4 months of PRRT was 56% (partial remission, 12%; stable disease, 44%). The disease control rate (72%) was higher by molecular response evaluation. Median OS and PFS were 63.7 and 10.1 months, respectively. The annual OS rates were 84% at 1 year, 65% at 3 years, 57% at 5 years, and 18% at 10 years. The annual PFS rates were 42% at 1 year, 21% at 2 years, and 13% at 5 years. Patients with bone metastases had poorer OS and PFS than those without metastases (median OS, 58.7 vs 92.3 months [P = 0.035; hazard ratio, 2.7; 95% confidence interval, 0.92-7.84]; median PFS, 8.5 vs 12.8 months [P = 0.592; hazard ratio, 1.2; 95% confidence interval, 0.56-2.76]). CONCLUSIONS: Peptide receptor radionuclide therapy was well tolerated and effective in patients with advanced, aggressive MTC. Bone metastasis was an independent adverse prognostic factor for OS.


Asunto(s)
Tumores Neuroendocrinos , Compuestos Organometálicos , Neoplasias de la Tiroides , Masculino , Humanos , Femenino , Adulto , Persona de Mediana Edad , Octreótido/efectos adversos , Radioisótopos de Itrio , Tumores Neuroendocrinos/patología , Progresión de la Enfermedad , Neoplasias de la Tiroides/radioterapia , Neoplasias de la Tiroides/tratamiento farmacológico , Receptores de Péptidos/uso terapéutico , Compuestos Organometálicos/efectos adversos , Estudios Retrospectivos
5.
Front Endocrinol (Lausanne) ; 14: 1270421, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38317712

RESUMEN

Introduction: Peptide receptor radionuclide therapy (PRRT) is a treatment option for well-differentiated, somatostatin receptor positive, unresectable or/and metastatic neuroendocrine tumors (NETs). Although high disease control rates seen with PRRT a significant number NET patients have a short progression-free interval, and currently, there is a deficiency of effective biomarkers to pre-identify these patients. This study is aimed at determining the prognostic significance of biomarkers on survival of patients with NETs in initial PRRT treatment. Methodology: We retrospectively analyzed 51 patients with NETs treated with PRRT at the Department for nuclear medicine, University Clinical Center Kragujevac, Serbia, with a five-year follow-up. Eligible patients with confirmed inoperable NETs, were retrospectively evaluated hematological, blood-based inflammatory markers, biochemical markers and clinical characteristics on disease progression. In accordance with the progression og the disease, the patients were divided into two groups: progression group (n=18) and a non-progression group (n=33). Clinical data were compared between the two groups. Results: A total of 51 patients (Md=60, age 25-75 years) were treated with PRRT, of whom 29 (56.86%) demonstrated stable disease, 4 (7.84%) demonstrated a partial response, and 14 (27.46%) demonstrated progressive disease and death was recorded in 4 (7.84%) patients. The mean PFS was a 36.22 months (95% CI 30.14-42.29) and the mean OS was 44.68 months (95% CI 37.40-51.97). Univariate logistic regression analysis displayed that age (p<0.05), functional tumors (p<0.05), absolute neutrophil count (p<0.05), neutrophil-lymphocyte ratio-NLR (p<0.05), C-reactive protein-CRP (p<0.05), CRP/Albumin (p<0.05), alanine aminotransferase-ALT (p<0.05), were risk factors for disease progression. Multivariate logistic regression analysis exhibited that functional tumors (p<0.001), age (p<0.05), CRP (p<0.05), and ALT (p<0.05), were independent risk factors for the disease progression in patients with NETs. Tumor functionality was the most powerful prognostic factor. The median PFS (11.86 ± 1.41 vs. 43.38 ± 3.16 months; p=0.001) and OS (21.81 ± 2.70 vs 53.86 ± 3.70, p=0.001) were significantly shorter in patients with functional than non-functional NETs respectively. Conclusion: The study's results suggest that tumor functionality, and certain biomarkers may serve as prognostic survival indicators for patients with NETs undergoing PRRT. The findings can potentially help to identify patients who are at higher risk of disease progression and tailor treatment strategies accordingly.


Asunto(s)
Tumores Neuroendocrinos , Octreótido , Humanos , Adulto , Persona de Mediana Edad , Anciano , Octreótido/uso terapéutico , Estudios Retrospectivos , Serbia/epidemiología , Tumores Neuroendocrinos/tratamiento farmacológico , Radioisótopos/uso terapéutico , Progresión de la Enfermedad , Biomarcadores , Receptores de Péptidos/uso terapéutico
6.
Anticancer Res ; 42(10): 4679-4687, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36191971

RESUMEN

BACKGROUND/AIM: Thrombomodulin™ has cytoprotective and anti-inflammatory function by interacting with G-protein coupled receptor 15 (GPR15). Recombinant TM (rTM), which comprises the extracellular regions of TM, is approved for treatment of disseminated intravascular coagulation. We investigated the anti-tumor effect of rTM for pancreatic ductal adenocarcinoma (PDAC) through GPR15. MATERIALS AND METHODS: We evaluated the expression of GPR15 in human PDAC cell lines and the anti-tumor effect and signals of rTM in vitro and in vivo. To test whether GPR15 would be responsible for the inhibition of cell proliferation by rTM, we evaluated the cell viability of the GPR15 knockdown cells treated with rTM using GPR15-targeting siRNA. RESULTS: We identified PDAC cell lines with GPR15 expression and discovered that rTM inhibited tumor growth and enhanced the effects of gemcitabine (GEM) for the PDAC cell line in a GPR15-dependent manner. Furthermore, we showed that rTM inhibited nuclear factor-kappaB (NF-[Formula: see text]B) and extracellular signal-regulated kinase (ERK) activation through interactions with GPR15. CONCLUSION: We demonstrated that rTM had anti-tumor effect and enhancement of cytotoxic effect of GEM for PDAC cells by inhibiting NF-[Formula: see text]B and ERK activation via GPR15 and suggest that rTM is a potential therapeutic option for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Antiinflamatorios/uso terapéutico , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Quinasas MAP Reguladas por Señal Extracelular , Humanos , FN-kappa B/metabolismo , Neoplasias Pancreáticas/patología , ARN Interferente Pequeño , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/uso terapéutico , Trombomodulina/genética , Trombomodulina/uso terapéutico , Gemcitabina , Neoplasias Pancreáticas
7.
Eur J Clin Pharmacol ; 78(9): 1365-1376, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35729340

RESUMEN

OBJECTIVE: Calcitonin gene-related peptide (CGRP) receptor antagonists have been suggested as novel treatments for acute migraine. This study aimed to use meta-analysis to compare the safety and tolerability of five existing oral CGRP receptor antagonists (BI44370TA, MK-3207, rimegepant, telcagepant, and ubrogepant) with that of a placebo or triptans against acute migraine. METHODS: Five prominent databases were searched to identify randomized controlled trials on this topic. The primary safety outcomes of interest were any adverse events (AEs) and treatment-related adverse events (TRAEs), and secondary outcomes were individual events, namely diarrhea, dizziness, dry mouth, fatigue, nausea, paresthesia, somnolence, upper abdominal pain, and vomiting. RESULTS: Fifteen studies met the eligibility criteria and were examined in detail. Although, compared to placebo, oral CGRP receptor antagonists significantly increased the incidence of any AEs (risk ratio [RR] = 1.15; 95% confidence interval [CI] = 1.07-1.23), there was no difference in the incidence of TRAEs (RR = 1.18; 95% CI = 1.00-1.38). Moreover, CGRP receptor antagonists were safer than triptans with respect to primary safety outcomes, such as any AEs (RR = 0.78; 95% CI = 0.63-0.98) and TRAEs (RR = 0.68; 95% CI = 0.58-0.79). CONCLUSION: Despite oral CGRP receptor antagonists posing a significantly higher risk of AEs when compared to placebo, CGRP receptor antagonists have a favorable safety profile compared to triptans. Our findings inform strategies to enhance safety and tolerability in the treatment of acute migraine.


Asunto(s)
Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina , Trastornos Migrañosos , Calcitonina/uso terapéutico , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/efectos adversos , Humanos , Trastornos Migrañosos/tratamiento farmacológico , Receptores de Péptidos/uso terapéutico , Triptaminas/efectos adversos
8.
Adv Pharmacol ; 94: 183-211, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35659372

RESUMEN

The pregnancy related hormone relaxin is produced throughout the reproductive system. However, relaxin also has important cardiovascular effects as part of the adaptation that the cardiovascular system undergoes in response to the extra demands of pregnancy. These effects are primarily mediated by the relaxin family peptide receptor 1, which is one of four known relaxin receptors. The effects of relaxin on the cardiovascular system during pregnancy, as well as its anti-fibrotic and anti-inflammatory properties, have led to extensive studies into the potential of relaxin therapy as an approach to treat heart failure. Cardiomyocytes, cardiac fibroblasts, and endothelial cells all possess relaxin family peptide receptor 1, allowing for direct effects of therapeutic relaxin on the heart. Many pre-clinical animal studies have demonstrated a beneficial effect of exogenous relaxin on adverse cardiac remodeling including inflammation, fibrosis, cardiomyocyte hypertrophy and apoptosis, as well as effects on cardiac contractile function. Despite this, clinical studies have yielded disappointing results for the synthetic seralaxin, even though seralaxin was well tolerated. This article will provide background on relaxin in the context of normal physiology, as well as the role of relaxin in pregnancy-related adaptations of the cardiovascular system. We will also present evidence from pre-clinical animal studies that demonstrate the potential benefits of relaxin therapy, as well as discussing the results from clinical trials. Finally, we will discuss possible reasons for the failure of these clinical trials as well as steps being taken to potentially improve relaxin therapy for heart failure.


Asunto(s)
Cardiopatías , Insuficiencia Cardíaca , Hipertensión , Relaxina , Animales , Células Endoteliales , Femenino , Fibrosis , Cardiopatías/inducido químicamente , Cardiopatías/tratamiento farmacológico , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Embarazo , Receptores de Péptidos/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Relaxina/efectos adversos
9.
Zhonghua Yi Xue Za Zhi ; 102(14): 1034-1038, 2022 Apr 12.
Artículo en Chino | MEDLINE | ID: mdl-35399024

RESUMEN

The clinical data of 14 patients with neuroendocrine tumors who received Peptide Receptor Radionuclide Therapy (PRRT) from December 2018 to May 2021 were retrospectively analyzed. Among them, 2 patients demonstrated proprogressive disease, 2 patients had partial response, and 10 patients had stable disease. Grade 1-2 myelosuppression occurred in 5 patients. and 1 patient became grade 3 myelosuppression,which recovered to grade 2 after symptomatic treatment. No grade 2 or higher treatment-related renal toxicity was observed in any of the patients. PRRT is efficacy and no significant side effects for unresectable metastatic neuroendocrine tumors.


Asunto(s)
Tumores Neuroendocrinos , Humanos , Tumores Neuroendocrinos/radioterapia , Octreótido/uso terapéutico , Radioisótopos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Estudios Retrospectivos
10.
Exp Cell Res ; 413(2): 113078, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35189107

RESUMEN

Although MET tyrosine kinase inhibitors (TKIs) are generally effective against non-small cell lung carcinoma (NSCLC) with MET exon 14 skipping mutations (METΔex14), resistance to MET TKIs can occur, indicating the need to develop other therapeutic options. We found that Hs-746 T cells, which harbor METΔex14 plus amplification, were able to survive and grow in the absence of MET signaling, exhibiting primary resistance to MET TKIs. We also found a moderately positive correlation between MET and anthrax toxin receptor 2 (ANTXR2) mRNA expression in NSCLC cell lines using data from the Cancer Dependency Map database. As expected, Hs-746 T cells were positive for ANTXR2 expression. We used an antibody-drug conjugate (ADC) analog in the form of an anti-ANTXR2 monoclonal antibody, H8R23, conjugated to DT3C recombinant protein which consists of diphtheria toxin (DT) lacking the receptor-binding domain but containing the C1, C2, and C3 domains of streptococcal protein G (3C). H8R23-DT3C conjugates, which function in vitro like an ADC, induced Hs-746 T cells to undergo apoptosis, resulting in decreased viability. These findings collectively suggest that an ADC targeting ANTXR2 could be effective for the treatment of METΔex14-positive NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-met/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Exones/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Péptidos/genética , Receptores de Péptidos/uso terapéutico
11.
JAMA Netw Open ; 5(1): e2144170, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-35044469

RESUMEN

Importance: Despite the benefit of peptide receptor radionuclide therapy (PRRT) for patients with well-differentiated neuroendocrine tumors (WD NETs), no clinical metric to anticipate benefit from the therapy for individual patients has been previously defined. Objective: To assess whether the prognostic ability of the clinical score (CS) could be validated in an external cohort of patients with WD NETs. Design, Setting, and Participants: This multicenter cohort study's analysis included patients with WD NETs who were under consideration for peptide receptor radionuclide therapy (PRRT) with lutetium-177 (177Lu)-dotatate between March 1, 2016, and March 17, 2020. The original cohort included patients from Vanderbilt-Ingram Cancer Center. The validation cohort included patients from Ochsner Medical Center, Markey Cancer Center, and Rush Medical Center. Patients with paragangliomas, pheochromocytomas and neuroblastomas were excluded. Statistical analysis was performed from June to November 2021. Exposures: PRRT with 177Lu-dotatate or alternate therapies such as everolimus, sunitinib, or capecitabine plus temozolomide. Main Outcomes and Measures: The primary outcome was progression-free survival (PFS) and was estimated by the Kaplan-Meier method; a Cox proportional-hazards model adjusting for primary tumor site, tumor grade, and number of PRRT doses administered was used to analyze association between CS and outcomes. Results: A total of 126 patients (median age [IQR] age: 63.6 [52.9-70.7] years; 64 male individuals) were included in the validation cohort, and the combined cohort (validation and original cohorts combined) had a total of 248 patients (median [IQR] patient age: 63.3 [53.3-70.3] years; 126 male individuals). In the validation cohort, on multivariable analysis, for each 2-point increase in CS, PFS decreased significantly (hazard ratio, 2.61; 95% CI, 1.64-4.16). After finding an association of the CS with PFS in the validation cohort, the original and validation cohorts were combined into the cohort for this analysis. On multivariable analysis, for each 2-point increase in CS, PFS decreased significantly (hazard ratio, 2.52; 95% CI, 1.89-3.36). Conclusions and Relevance: Increases in CS were associated with worsening PFS in the validation cohort, validating findings from the original cohort. These findings suggest that the CS, to our knowledge, represents the first clinical metric to estimate anticipated benefit from PRRT for patients with WD NETs and may be a clinical tool for patients being considered for PRRT.


Asunto(s)
Tumores Neuroendocrinos/mortalidad , Tumores Neuroendocrinos/radioterapia , Radioisótopos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Índice de Severidad de la Enfermedad , Anciano , Estudios de Cohortes , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Compuestos Organometálicos/uso terapéutico , Valor Predictivo de las Pruebas , Pronóstico , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Cintigrafía , Resultado del Tratamiento
12.
J Nucl Med ; 63(3): 396-398, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34475234

RESUMEN

For patients with Merkel cell carcinoma (MCC) who are refractory to immune checkpoint inhibition (ICI), treatment options are limited. Few cases of MCCs have been reported to show responses to peptide receptor radionuclide therapy (PRRT). A combination of PRRT and ICI has not been reported in MCC to date. A patient with metastatic MCC, who was resistant to first-line avelumab and acquired resistance to ipilimumab/nivolumab (IPI/NIVO) with additional radiotherapy, presented with multiple distant metastases. After confirmation of SSTR expression, treatment was continued with an additional 4 doses of IPI/NIVO combined with 2 cycles of PRRT. Treatment was well tolerated, with transient hemotoxicity and mild nausea. Restaging after 3 mo demonstrated an exceptional response. This case demonstrates the feasibility of combined treatment with IPI/NIVO and PRRT as an option for MCC patients progressing under ICI. Prospective evidence confirming the additive value of combining ICI and radionuclide therapy in a larger cohort is needed.


Asunto(s)
Carcinoma de Células de Merkel , Radioisótopos , Neoplasias Cutáneas , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células de Merkel/tratamiento farmacológico , Carcinoma de Células de Merkel/etiología , Carcinoma de Células Renales/tratamiento farmacológico , Femenino , Humanos , Inmunoterapia , Ipilimumab/uso terapéutico , Neoplasias Renales/patología , Masculino , Nivolumab/uso terapéutico , Estudios Prospectivos , Radioisótopos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/terapia
13.
J Hepatobiliary Pancreat Sci ; 29(4): 487-499, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34907652

RESUMEN

BACKGROUND: The present prospective phase 1/2 study aimed to elucidate the efficacy and safety of 177 Lu-DOTATATE (four cycles of 7.4 GBq) in Japanese patients with unresectable, progressive neuroendocrine tumors (NETs). METHODS: From April 2018 to October 2020, 15 patients with advanced NETs (five midgut, eight pancreatic, and two lung NETs) were enrolled. Objective response rate (ORR), progression-free survival (PFS), and adverse events (AEs) were evaluated. Pharmacokinetics and dosimetry were also evaluated in three midgut patients. RESULTS: The mean absorbed doses of 177 Lu-DOTATATE to the kidneys (20.7 Gy/29.6 GBq) and the bone marrow (0.631 Gy/29.6 GBq) were within the radiation tolerance doses. The ORR of the whole population was 53% (90% CI, 30%-76%). ORRs of the midgut and non-midgut NETs were 60% (90% CI, 19%-92%) and 50% (90% CI, 22%-78%), respectively. There was no difference in the maximum reduction rate of the sum of the target lesion diameters between patients with midgut and non-midgut NET. The median PFS was not reached; the PFS rate at 52 weeks was 80% (90% CI, 56.1%-91.7%). AEs of Grade 3 or higher were lymphopenia (47%) and leukopenia (7%). CONCLUSION: 177 Lu-DOTATATE demonstrated remarkable tumor shrinkage and tolerability in Japanese patients with advanced NETs.


Asunto(s)
Tumores Neuroendocrinos , Humanos , Japón , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/radioterapia , Tomografía de Emisión de Positrones , Estudios Prospectivos , Radioisótopos/efectos adversos , Cintigrafía , Radiofármacos/efectos adversos , Radiofármacos/farmacocinética , Receptores de Péptidos/uso terapéutico
14.
J Clin Endocrinol Metab ; 106(12): e4903-e4916, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34379772

RESUMEN

CONTEXT: Metastatic medullary thyroid cancer (MTC) is a rare malignancy with minimal treatment options. Many, but not all, MTCs express somatostatin receptors. OBJECTIVE: Our aim was to explore the role of 68Ga-DOTA-somatostatin analogue (SSA) positron emission tomography (PET)/computed tomography (CT) in patients with metastatic MTC and to determine their eligibility for peptide receptor radionuclide therapy (PRRT). METHODS: We retrospectively identified patients with metastatic MTC who had 68Ga-DOTA-SSA PET/CT at 5 centers. We collected characteristics on contrast-enhanced CT, 68Ga-DOTA-SSA and 18F-FDG PET/CT. The efficacy of PRRT was explored in a subgroup of patients. Kaplan-Meier analysis was used to estimate time to treatment failure (TTF) and overall survival (OS). RESULTS: Seventy-one patients were included (10 local recurrence, 61 distant disease). Of the patients with distant disease, 16 (26%) had ≥50% of disease sites with tracer avidity greater than background liver, including 10 (10/61, 16%) with >90%. In 19 patients with contemporaneous contrast-enhanced CT, no disease regions were independently identified on 68Ga-DOTA-SSA PET/CT. Thirty-five patients had an 18F-FDG PET/CT, with 18F-FDG positive/68Ga-DOTA-SSA negative metastases identified in 15 (43%). Twenty-one patients had PRRT with a median TTF of 14 months (95% CI 8-25) and a median OS of 63 months (95% CI 21-not reached). Of the entire cohort, the median OS was 323 months (95% CI 152-not reached). Predictors of poorer OS included a short calcitonin doubling-time (≤24 months), strong 18F-FDG avidity, and age ≥60 years. CONCLUSIONS: The prevalence of high tumor avidity on 68Ga-DOTA-SSA PET/CT is low in the setting of metastatic MTC; nevertheless, PRRT may still be a viable treatment option in select patients.


Asunto(s)
Carcinoma Neuroendocrino/radioterapia , Compuestos Organometálicos/uso terapéutico , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Receptores de Péptidos/uso terapéutico , Receptores de Somatostatina/uso terapéutico , Somatostatina/química , Neoplasias de la Tiroides/radioterapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Neuroendocrino/diagnóstico por imagen , Carcinoma Neuroendocrino/secundario , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/secundario , Adulto Joven
15.
Curr Treat Options Oncol ; 22(5): 43, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33786683

RESUMEN

OPINION STATEMENT: Neuroendocrine neoplasms (NENs) constitute a heterogenous group of malignancies. Translational research into NEN cell biology is the cornerstone for drug development strategies in this field. Somatostatin receptor type 2 (SSTR2) expression is the hallmark of well-differentiated neuroendocrine tumors (NETs). Somatostatin analogs and peptide receptor radionuclide therapy (PRRT) form the basis of anti-SSTR2 treatment onto new combination strategies, antibody-drug conjugates and bispecific antibodies. Classical pathways involved in NET development (PI3K-Akt-mTOR and antiangiogenics) are reviewed but new potential targets for NET treatment will be explored. Epigenetic drugs have shown clinical activity in monotherapy and preclinical combination strategies are more than attractive. Immunotherapy has shown opposite results in different NEN settings. Although the NOTCH pathway has been targeted with disappointing results, new strategies are being developed. Finally, after years of solid preclinical evidence on different genetically engineered oncolytic viruses, clinical trials for refractory NET patients are now ongoing.


Asunto(s)
Desarrollo de Medicamentos , Tumores Neuroendocrinos/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Desarrollo de Medicamentos/tendencias , Epigénesis Genética/efectos de los fármacos , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Virus Oncolíticos , Inhibidores de Proteínas Quinasas/uso terapéutico , Radioisótopos/química , Radioisótopos/uso terapéutico , Receptores de Péptidos/química , Receptores de Péptidos/uso terapéutico , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico
16.
Neuroendocrinology ; 111(11): 1086-1098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33744879

RESUMEN

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are the most common form of neuroendocrine neoplasia, but there is no current consensus for the sequencing of approved therapies, particularly with respect to peptide receptor radionuclide therapy (PRRT). This comprehensive review evaluates the data supporting approved therapies for GEP-NETs and recommendations for therapeutic sequencing with a focus on how PRRT currently fits within sequencing algorithms. The current recommendations for PRRT sequencing restrict its use to metastatic, inoperable, progressive midgut NETs; however, this may change with emerging data to suggest that PRRT might be beneficial as neoadjuvant therapy for inoperable tumors, is more tolerable than other treatment modalities following first-line standard dose somatostatin analogs, and can be used as salvage therapy after disease relapse following prior successful cycles of PRRT. PRRT has also been shown to reduce tumor burden, improve quality of life, and prolong the time to disease progression in a broad spectrum of patients with GEP-NETs. As the various potential benefits of PRRT in GEP-NET therapy continues to expand, it is necessary to review and critically evaluate our treatment algorithms for GEP-NETs.


Asunto(s)
Neoplasias Intestinales/terapia , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/terapia , Radioisótopos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Neoplasias Gástricas/terapia , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/radioterapia , Neoplasias Intestinales/cirugía , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/radioterapia , Tumores Neuroendocrinos/cirugía , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/cirugía , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/radioterapia , Neoplasias Gástricas/cirugía
17.
Cancer Treat Rev ; 93: 102141, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33418096

RESUMEN

BACKGROUND: This review and meta-analysis examined published evidence of peptide receptor radionuclide therapy (PRRT) re-treatment efficacy and safety in patients with advanced neuroendocrine tumors (NETs). METHODS: Embase, MEDLINE, MEDLINE In-Progress, and Cochrane CENTRAL were searched (database inception-present) to identify evidence of efficacy and safety of PRRT re-treatment in adults with NETs previously treated with 177Lu- and/or 90Y-PRRT. Progression-free survival (PFS), overall survival (OS), disease control rate (DCR) from time of re-treatment were assessed. Data were pooled using medians and variance for time-to-event outcomes and inverse-variance weighted proportions (Freeman-Tukey method) for binary outcomes. RESULTS: Of 567 studies screened, 13 reported re-treatment efficacy outcomes. In random-effects meta-analyses of 177Lu-PRRT re-treatment, median PFS (N = 7 studies [414patients]) was 12.52 months (95% CI 9.82-15.22) with moderate heterogeneity across studies (I2 = 50.5%), median OS (N = 2 [194 patients]) was 26.78 months (95% CI 18.73-34.83) with moderate-to-high heterogeneity (I2 = 57.5%), and DCR (N = 8 [347 patients]) was 71% (95% CI 66-75) with high heterogeneity (I2 = 81.5%). PFS was similar with either 177Lu-PRRT re-treatment alone or in combination with 90Y-PRRT. Grade 3/4 adverse events occurred in 5% (95% CI 2-8) of patients receiving 177Lu-PRRT re-treatment (N = 5 [271 patients]) with few grade 3/4 renal toxicities (0% [95% CI 0-1]). Pooled myelodysplastic syndrome and acute myeloid leukemia incidence was 0% (95%CI 0-2). CONCLUSION: Re-treatment with 177Lu-PRRT provided encouraging median PFS in patients with NETs with a safety profile similar to initial PRRT.


Asunto(s)
Tumores Neuroendocrinos/radioterapia , Radiofármacos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Humanos , Tumores Neuroendocrinos/mortalidad , Tumores Neuroendocrinos/patología , Supervivencia sin Progresión , Radioterapia , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
18.
Curr Opin Oncol ; 33(1): 33-39, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33093336

RESUMEN

PURPOSE OF REVIEW: Pheochromocytomas and paragangliomas are rare tumors arising, respectively, from the adrenal medulla and extra-adrenal sympathetic or parasympathetic paraganglia. The main therapeutic objectives in case of metastatic disease are the reduction of tumor burden and the control of symptoms resulting from excessive catecholamine secretion. Treatment choices constitute not only a wait and see attitude, locoregional approaches, chemotherapy regiments but also radiopharmaceutical agents, and they should be discussed in a specialized multidisciplinary board. This review will briefly discuss the radiopharmaceutical modalities in patients with pheochromocytomas and paragangliomas (I-MIBG and PRRT). RECENT FINDINGS: I-MIBG (Azedra) has received FDA approval for patients with iobenguane-scan-positive, unresectable, locally advanced or metastatic pheochromocytomas and paragangliomas who require systemic anticancer therapy, whereas peptide receptor radionuclide therapy using radiolabelled somatostatin analogues is currently performed in compassionate use, with very promising results. No prospective head-to-head comparison between the modalities has been conducted to date. SUMMARY: Promising results have been reported for both radiopharmaceutical agents, mostly in the setting of retrospective series. No prospective head-to-head comparison between the modalities is yet available.


Asunto(s)
3-Yodobencilguanidina/uso terapéutico , Neoplasias de las Glándulas Suprarrenales/radioterapia , Radioisótopos de Yodo/uso terapéutico , Paraganglioma/radioterapia , Feocromocitoma/radioterapia , Radiofármacos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Humanos
19.
Radiology ; 298(2): 261-274, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33231532

RESUMEN

Lutetium 177 (177Lu) DOTA-0-Tyr3-Octreotate (DOTATATE) peptide receptor radionuclide therapy (PRRT) is an effective treatment for advanced gastroenteropancreatic neuroendocrine tumors. This review presents a clinical practice workflow that has been successful since 177Lu DOTATATE PRRT was approved by the U.S. Food and Drug Administration. The workflow relies heavily on the input of a multidisciplinary team and involves a nuclear medicine consultation service, tumor board, and specific preparations in advance of therapy and day-of-therapy procedures. A systematic checklist designed to ensure appropriate selection of treatment candidates and identification of any concerns to address to safely administer PRRT is provided. All patients were evaluated with gallium 68 DOTATATE PET/CT, and in cases of high-grade tumors, they were also evaluated with fluorine 18 fluorodeoxyglucose PET/CT, with imaging findings reviewed as part of the systematic checklist before PRRT. Adverse effects are discussed and imaging follow-up regimens are reviewed, including alternative diagnostic contrast materials. Approaches to multiple challenging patient scenarios are illustrated through case examples. Finally, alternative theranostic radionuclides and treatment strategies are discussed.


Asunto(s)
Neoplasias Intestinales/radioterapia , Tumores Neuroendocrinos/radioterapia , Octreótido/análogos & derivados , Compuestos Organometálicos/uso terapéutico , Neoplasias Pancreáticas/radioterapia , Radiofármacos/uso terapéutico , Receptores de Péptidos/uso terapéutico , Neoplasias Gástricas/radioterapia , Humanos , Neoplasias Intestinales/diagnóstico por imagen , Imagen por Resonancia Magnética , Tumores Neuroendocrinos/diagnóstico por imagen , Octreótido/uso terapéutico , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Gástricas/diagnóstico por imagen , Tomografía Computarizada por Rayos X
20.
Clin J Oncol Nurs ; 24(2): 129-133, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32195999

RESUMEN

Neuroendocrine tumors (NETs) are comprised of biologically diverse neoplasms. The presence of systemic symptoms is dependent on NET location and differentiation. New treatment modalities have become available, offering patients improved symptom management and survival. Advanced practice RNs (APRNs) provide direct care to and coordination of treatment for patients with NETs, including treatment of somatostatin receptor-positive disease with lutetium Lu 177 dotatate (Lutathera®) peptide receptor radionuclide therapy.


Asunto(s)
Complejos de Coordinación/uso terapéutico , Neoplasias Gastrointestinales/terapia , Tumores Neuroendocrinos/terapia , Octreótido/análogos & derivados , Receptores de Péptidos/uso terapéutico , Adulto , Femenino , Humanos , Lutecio , Enfermeras y Enfermeros , Octreótido/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...